Nanotherapy has emerged as an improved anticancer therapeutic strategy to circumvent the harmful side effects of chemotherapy. It has been proven to be beneficial to offer multiple advantages, including their capacity to carry different therapeutic agents, longer circulation time and increased therapeutic index with reduced toxicity. Over time, nanotherapy evolved in terms of their designing strategies like geometry, size, composition or chemistry to circumvent the biological barriers. Multifunctional nanoscale materials are widely used as molecular transporter for delivering therapeutics and imaging agents. Nanomedicine involving multi-component chemotherapeutic drug-based combination therapy has been found to be an improved promising approach to increase the efficacy of cancer treatment. Next-generation nanomedicine has also utilized and combined immunotherapy to increase its therapeutic efficacy. It helps in targeting tumor immune response sparing the healthy systemic immune function. In this review, we have summarized the progress of nanotechnology in terms of nanoparticle designing and targeting cancer. We have also discussed its further applications in combination therapy and cancer immunotherapy. Integrating patient-specific proteomics and biomarker based information and harnessing clinically safe nanotechnology, the development of precision nanomedicine could revolutionize the effective cancer therapy.

Cancer remains the most fatal and life-threatening disease worldwide, causing an estimated 19.3 million new cases and 10.0 million deaths in 2020 [1]. Current therapeutic approaches are not found to completely cure the advanced cancer having distant organ metastases. Treatment of cancer follows several approaches, including locoregional (surgery and radiation therapy), chemotherapy, endocrine (hormone) therapy, targeted therapy, etc. Such therapeutic approaches also possess certain disadvantages such as fatigue, numbness, nail changes, hair loss, loss of appetite, mouth sores, nausea, weight changes, vomiting, diarrhoea, heart damage, etc [2–6]. In spite of its huge potential, chemotherapy remains disadvantageous in having off-target side effects and non-specific delivery [7]. Nanotechnology has become a promising approach to overcome this limitation. The nano-sized materials provide opportunities for their use in diagnosing, monitoring, controlling, preventing and treating diseases [8]. The concept of nanoparticles was first adapted by Nobel laureate Richard P. Feynman in his famous lecture entitled “There’s plenty of room at the bottom” in December 1959 [9]. Nanotherapy is the therapeutic strategy harboring nanoparticles ranging from 10 to 100 nm for intravenous delivery [10,11]. Nanoparticles below 10 nm diameter are prone to renal clearance [11]. Widely used examples of nanoparticles having anticancer efficacy are Doxil, a liposomal formulation of Doxorubicin and Abraxane, an albumin-bound nanoparticle of paclitaxel [12]. These drugs also received approval from U.S. Food and Drug Administration (FDA). Apart from that, several other nanotechnology platforms like organic, inorganic and organometallic nanoparticles have also been used over the past two decades for therapeutic, diagnostic and theranostic purposes [13]. Nanoparticles can be easily custom-tailored and advantageous in having the following features: (1) carry a high payload of biologically active drug [11,14,15], (2) protects the drug from degradation, (3) contain large surface area to accommodate multiple targeting ligands (4) larger surface to interact with multiple types of drug molecules, (5) controlled release profile of drug and (6) potential to bypass multidrug resistant mechanism [11,16,17].

Nanotechnology also undergoes several barriers that hinder its successful translation into the clinic. Various biological barriers limit the functionality of the nanoparticles and their clinical outcome. Nanoparticles get easily phagocytosed and degraded by the macrophages of the liver, spleen, lungs, lymph nodes and skin [13]. According to recent statistics, 0.7% of the total injected dose can get targeted to tumor. The primary reason for this failure is the formation of protein corona (proteins adsorbed on the nanoparticle from plasma and/or intracellular fluid) around nanoparticles and their subsequent opsonization (the process where opsonins or extracellular proteins bind to the surface of the nanoparticles, causing the degradation of nanoparticles by phagocytes) and eventual phagocytosis [10,13]. High encapsulation efficiency of a drug does not always lead to high therapeutic potential. Again, hydrophobic drugs which precipitate over time also do not show significant efficacy [10]. Therefore, designing a potent yet stable nanoparticle has always been challenging. In the following section, we will focus on the strategies acquired by the researchers in designing effective nanoparticles to aid anticancer therapy and we will also describe the progress made so far with nanotherapy.

By designing suitable combinatorial nanotherapeutics, one can achieve targeted delivery, reduce side effects of free drugs, delay in developing drug resistance and accomplish synergistic drug interactions at low doses [18]. Oncogenic signaling pathways can also be targeted using nanoparticles to selectively target the tumor cells without causing systemic toxicity. Moreover, nanomedicine mediated anti-angiogenic drug delivery makes them more reachable towards tumor vasculature. Furthermore, nanoparticles potentiate the gene therapy to introduce therapeutic nucleic acids into target cells to achieve curative response in cancer patients [19]. Recently, nanotherapy has also been used to deliver immunotherapeutic drugs in combination with conventional therapeutic modalities (e.g., chemotherapy, RNAi therapy, photothermal, photodynamic and radiotherapy) to increase anti-tumor efficacy [20]. Minimally invasive photothermal therapy allows killing of cancer cells by heat generated upon their exposure to the near infrared (NIR) light [20]. Similarly, photodynamic therapy utilizes photosensitizers which upon exposure to light releases reactive oxygen species and induce cellular toxicity toward tumors [20]. Radiotherapy induced radiation damage of cancer cells is widely used in cancer treatment, which allows curative treatment of 40% patients out of >50% of the patients with cancer treated by radiotherapy [20,21]. Cancer immunotherapy has gained tremendous attention for providing long-term treatment in cancer, as it facilitates immunological memory induced delay in cancer remission. One of the key challenges of implementing immunotherapy is off-target responses. Nanotherapy has potential to overcome this challenge by delivering these immunotherapeutic drugs to desired target sites [22]. In this review, we have discussed the potential applications and the strength of nanotherapy by combining therapies like chemotherapy, gene therapy and immunotherapy. The central theme of this review is on engineering multifunctional cancer nanotherapy as described in Figure 1.

Overview of the basis of multifunctional cancer nanotherapy and its potential applications

Figure 1
Overview of the basis of multifunctional cancer nanotherapy and its potential applications

The figure describes the targeting strategies involved in nanotherapy, the composition of nanoparticles, types of cargo and its further applications in combination therapies.

Figure 1
Overview of the basis of multifunctional cancer nanotherapy and its potential applications

The figure describes the targeting strategies involved in nanotherapy, the composition of nanoparticles, types of cargo and its further applications in combination therapies.

Close modal

Engineering multifunctional nanoparticle or precision nanomedicine still remains challenging. The primary aim of nanoparticle design is to transport therapeutic drugs and imaging agents. Such cargos are loaded on to nanoparticles either by chemical conjugation or by encapsulation [7]. Researchers found that the encapsulation technique remains more effective for clinical translation of a low potency drug than chemical conjugation. In order to produce efficacy, the drug should comprise at least 10% (wt/wt) of the entire nanoparticle composition [13]. While designing nanoparticles, certain criteria are needed to be taken into consideration, such as stability, pharmacological reasonability, pathophysiological suitability, etc [13]. Current development of cancer nanomedicine has provided answers to problems like (1) prolonged blood circulation to improve stability and bioavailability, (2) adequate tumor accumulation and (3) controlled drug release and uptake by tumor cells having a release profile for specific targeting [16,17].

Proteins present in biological fluids get adsorbed in the nanoparticles when administered. Such nanoparticle-protein complexes get easily recognized by macrophages and phagocytosed [10,23,24]. To overcome this difficulty, poly(ethylene glycol) (PEG) is often used to form a hydrophilic “stealth” [25]. PEGylation promotes solubilization of the nanoparticles, prevents opsonization and increases the half-life of the drug in the blood stream [26]. Doxil (liposomal doxorubicin) is a commonly used PEGylated nanoparticle. It shows ∼100 times longer half-life compared with free doxorubicin in circulation [27]. Current research has utilized nanotechnology to increase the solubility of potent but poorly soluble drugs such as cyclosporine, paclitaxel, amphotericin B, etc [28].

Nanoparticles possess unique properties due to their small size termed as “enhanced permeability and retention effect” (EPR effect) (Figure 2A) [29,30]. Harboring this phenomenon, nanoparticles extravasate through the leaky blood vessels (inter endothelial gaps as large as 500 nm) of the tumor vasculature and preferably accumulate at the tumor site. Due to poor lymphatic drainage, they retain at the tumor site. This phenomenon is also termed “passive targeting” (Figure 2A). Nanotherapeutics used for passive targeting are listed in Tables 1 and 11. However, targeting nanoparticles to the tumor tissue does not always remain successful due to variable vessel permeability [7].

Targeting strategies of nanoparticles

Figure 2
Targeting strategies of nanoparticles

Figure illustrating (A) the passive targeting (enhanced permeability and retention effect or EPR effect) and (B) the active targeting into a tumor. In passive targeting, nanoparticles extravasate through the leaky blood vessels of tumor vasculature having gap sizes of 100 nm to 2 μm. Due to poor lymphatic drainage, nanoparticles home at the tumor site. In active targeting, targeting ligands are attached to the nanoparticles that specifically target cancer's overexpressed receptors. The optimal size range to perform the EPR effect is 20–200 nm.

Figure 2
Targeting strategies of nanoparticles

Figure illustrating (A) the passive targeting (enhanced permeability and retention effect or EPR effect) and (B) the active targeting into a tumor. In passive targeting, nanoparticles extravasate through the leaky blood vessels of tumor vasculature having gap sizes of 100 nm to 2 μm. Due to poor lymphatic drainage, nanoparticles home at the tumor site. In active targeting, targeting ligands are attached to the nanoparticles that specifically target cancer's overexpressed receptors. The optimal size range to perform the EPR effect is 20–200 nm.

Close modal
Table 1
Nanotherapeutics used for passive targeting
NameFormulationDiameter (nm)CommentsRef.
SP1049C Pluronic micelle+DOX 22–27 Micelle nanoparticle [11,31
NK911 PEF-Asp micelle + DOX 40 Micelle nanoparticle [11,31
Doxil PEG-liposome +DOX 80–90 PEGylated liposome nanoparticle with a more extended time of circulation, less toxicity, prevents phagocytosis [11,31
Genexol-PM PEG- poly(l-lactic acid (PLA) micelle+paclitaxel 20–50 Micelle nanoparticle [11,31
Abraxane Albumin+paclitaxel 120 (May dissolve upon exposure to blood) Albumin nanoparticle [11,32
XYOTAX Poly-l-glutamic acid (PG) + paclitaxel Not reported Polymer nanoparticle [11,33
LE-SN-38 Liposome+SN-38 Not reported Liposome nanoparticle [11,34
CT-2106 PG+campothecin Not reported Polymer nanoparticle [11,35
IT-101 Cyclodextrin-containing polymer+campothecin 30–40 Polymer nanoparticle with extended circulation times [11,36
CCN Candesartan cilexetil loaded nanoemulsion 35.5 ± 5.9 Nanoemulsion formulation with increased aqueous solubility [37
NCS-DOX Nanocapsules with oily selol core and a shell of poly(methyl vinyl ether-co-maleic anhydride) + DOX 170 Poly(methyl vinyl ether-co-maleic anhydride) nanocapsules facilitate the co-delivery of drugs [38
NCI/NCa (DNase)-degradable DNA nanoclew embedded with an acid-responsive DNase I nanocapsule (NCa) + DOX 150–180 DNA-based nanoparticle [39
MWCNTs/DOX/TC TAT Chitosan functionalized multi walled carbon nanotube (MWCNT) + DOX 200–300 Multiwalled carbon nanotube-based nanosystem [40
NameFormulationDiameter (nm)CommentsRef.
SP1049C Pluronic micelle+DOX 22–27 Micelle nanoparticle [11,31
NK911 PEF-Asp micelle + DOX 40 Micelle nanoparticle [11,31
Doxil PEG-liposome +DOX 80–90 PEGylated liposome nanoparticle with a more extended time of circulation, less toxicity, prevents phagocytosis [11,31
Genexol-PM PEG- poly(l-lactic acid (PLA) micelle+paclitaxel 20–50 Micelle nanoparticle [11,31
Abraxane Albumin+paclitaxel 120 (May dissolve upon exposure to blood) Albumin nanoparticle [11,32
XYOTAX Poly-l-glutamic acid (PG) + paclitaxel Not reported Polymer nanoparticle [11,33
LE-SN-38 Liposome+SN-38 Not reported Liposome nanoparticle [11,34
CT-2106 PG+campothecin Not reported Polymer nanoparticle [11,35
IT-101 Cyclodextrin-containing polymer+campothecin 30–40 Polymer nanoparticle with extended circulation times [11,36
CCN Candesartan cilexetil loaded nanoemulsion 35.5 ± 5.9 Nanoemulsion formulation with increased aqueous solubility [37
NCS-DOX Nanocapsules with oily selol core and a shell of poly(methyl vinyl ether-co-maleic anhydride) + DOX 170 Poly(methyl vinyl ether-co-maleic anhydride) nanocapsules facilitate the co-delivery of drugs [38
NCI/NCa (DNase)-degradable DNA nanoclew embedded with an acid-responsive DNase I nanocapsule (NCa) + DOX 150–180 DNA-based nanoparticle [39
MWCNTs/DOX/TC TAT Chitosan functionalized multi walled carbon nanotube (MWCNT) + DOX 200–300 Multiwalled carbon nanotube-based nanosystem [40

In order to circumvent these barriers, researchers have taken advantage of conjugating tumor-targeting ligands to the nanoparticle surface resulting in “active targeting” (Figure 2B) [7,41]. Binding of ligand to exposed cell surface results in “receptor-mediated endocytosis”. However, receptor density plays a pivotal role in such targeting. Commonly used targeting agents are proteins (antibody and its fragments), ligands of the up-regulated receptors (peptides, carbohydrates and other small molecules) and nucleic acid aptamers [11]. Nanotherapeutics used for active targeting under clinical trial are mentioned in Tables 2 and 11. Peptides have been widely used as receptor targeting moieties for active targeting. Table 3 enlists an elaborate list of peptide-based targeting ligands which have already been reported by Gray and Brown [42].

Table 2
Antibody and peptide-based nanotherapeutics used for active targeting in the clinical trial
NameTargeting agentTherapeutic agentStatusCommentsRef.
Gemtuzumab ozogamicin (Mylotarg) Humanized anti-CD33 antibody Calicheamicin FDA approved Antibody-drug conjugate (ADC) [11,43
Denileukin diftitox (Ontak) Interleukin 2 (IL-2) Diphtheria toxin fragment FDA approved Recombinant fusion protein of IL-2 attached to diphtheria toxin fragments [11,44
Ibritumomab tiuxetan (Zevalin) Mouse anti-CD20 antibody 90Yttrium FDA approved Antibody–radioactive element conjugate [11,45
Tositumomab (Bexxar) Mouse anti-CD20 antibody 131Iodine FDA approved Antibody–radioactive element conjugate [11,45
FCE28069 (PK2) Galactose Doxorubicin Phase I clinical trial (stopped) A conjugate of HPMA copolymer, Doxorubicin and galactose [11,46
MCC-465 F(ab′)2 fragment of human antibody GAH Doxorubicin Phase I clinical trial Immunoliposome-encapsulated Doxorubicin (DXR) [11,47
MBP-426 Transferrin Oxaliplatin Phase I clinical trial Liposomal oxaliplatin suspension for injection [11,48
SGT-53 Antibody fragment to transferrin receptor Plasmid DNA with p53 gene Phase II clinical trial A complex of cationic liposome encapsulating p53 DNA sequence in a plasmid backbone [11,49
CALAA-01 Transferrin Small interfering RNA Phase I clinical trial Polymer-based nanoparticle having human transferrin protein targeting agent [11,50
BIND-014 Prostate-specific membrane antigen (PSMA)–targeted peptide (GDHSPFT, SHFSVGS and EVPRLSLLAVFL) Docetaxel Phase II clinical trail PLGA-PEG nanoparticle [51,52
NameTargeting agentTherapeutic agentStatusCommentsRef.
Gemtuzumab ozogamicin (Mylotarg) Humanized anti-CD33 antibody Calicheamicin FDA approved Antibody-drug conjugate (ADC) [11,43
Denileukin diftitox (Ontak) Interleukin 2 (IL-2) Diphtheria toxin fragment FDA approved Recombinant fusion protein of IL-2 attached to diphtheria toxin fragments [11,44
Ibritumomab tiuxetan (Zevalin) Mouse anti-CD20 antibody 90Yttrium FDA approved Antibody–radioactive element conjugate [11,45
Tositumomab (Bexxar) Mouse anti-CD20 antibody 131Iodine FDA approved Antibody–radioactive element conjugate [11,45
FCE28069 (PK2) Galactose Doxorubicin Phase I clinical trial (stopped) A conjugate of HPMA copolymer, Doxorubicin and galactose [11,46
MCC-465 F(ab′)2 fragment of human antibody GAH Doxorubicin Phase I clinical trial Immunoliposome-encapsulated Doxorubicin (DXR) [11,47
MBP-426 Transferrin Oxaliplatin Phase I clinical trial Liposomal oxaliplatin suspension for injection [11,48
SGT-53 Antibody fragment to transferrin receptor Plasmid DNA with p53 gene Phase II clinical trial A complex of cationic liposome encapsulating p53 DNA sequence in a plasmid backbone [11,49
CALAA-01 Transferrin Small interfering RNA Phase I clinical trial Polymer-based nanoparticle having human transferrin protein targeting agent [11,50
BIND-014 Prostate-specific membrane antigen (PSMA)–targeted peptide (GDHSPFT, SHFSVGS and EVPRLSLLAVFL) Docetaxel Phase II clinical trail PLGA-PEG nanoparticle [51,52
Table 3
Receptor targeting peptide sequences
ReceptorsCell linePeptide sequenceRef.
Met MDA-MB-231 YLFSVHWPPLKA [42
HER2/ErbB2 MDA-MB-231 KCCYSL [42
Transferrin MDA-MB-231, HeLa THRPPMWSPVWP [53
αvβ3 MDA-MB-231, HUVEC CDCRGDCFC [42
EGFR MDA-MB-468, MDA-MB-231 YHWYGYTPQNVI [42
IL-6 receptor B9 LSLITRL [42
Somatostatin receptor Type 2 (SSTR2) Breast, ovarian and cervical cancer cell lines fc[CFwKTC]T(ol) (Octreotide) (f : DPhenylalanine, w : DTryptophan, c : cyclic) [54,55
Ghrelin receptor (GnRH-R) Breast, lung, ovarian and prostate cancer cell lines pGlu-HWSYkLRPG-NH2 (pGlu : Pyroglutamic acid, k : DLysine) [54
Bombesin/Bn receptors Prostate, breast, pancreas and small cell lung cancer cell line yQWAV-βAla-HF-Nle-NH2 (y : DTyrosine, Nle : Norleucine, βAla : β-Alanine) [54,56
Vasoactive intestinal peptide receptors (VIP receptors) Breast, colon and endocrine cancer tumor cells HSDAVFTDNYTRLRKQMAVKKYLNSILN-NH2 [54
Neurotensin receptor 1 (NTSR1) Breast, colon and pancreatic cancer cells RRPYIL [54,57
CCK2R Liver, thyroid and pancreatic cancer cells eAYGWMDF-NH2 (e : DGlutamic acid) [54
MC1Melanoma cells Ac-Nle-DHfRWGK-NH2 (Ac : Acetyl, f : DPhenylalanine) [54,58
Human Y1 receptor (hY1R) Ewing sarcoma and breast cancer cell lines YPSKPDFPGEDAPAEDLARYYSALRHYINLITRPRY-NH2 [54
N-cadherin HUVEC SWTLYTPSGQSK [42
Carbonic anhydrase IX Renal cell carcinoma cell lines YNTNHVPLSPKY [42,59
ReceptorsCell linePeptide sequenceRef.
Met MDA-MB-231 YLFSVHWPPLKA [42
HER2/ErbB2 MDA-MB-231 KCCYSL [42
Transferrin MDA-MB-231, HeLa THRPPMWSPVWP [53
αvβ3 MDA-MB-231, HUVEC CDCRGDCFC [42
EGFR MDA-MB-468, MDA-MB-231 YHWYGYTPQNVI [42
IL-6 receptor B9 LSLITRL [42
Somatostatin receptor Type 2 (SSTR2) Breast, ovarian and cervical cancer cell lines fc[CFwKTC]T(ol) (Octreotide) (f : DPhenylalanine, w : DTryptophan, c : cyclic) [54,55
Ghrelin receptor (GnRH-R) Breast, lung, ovarian and prostate cancer cell lines pGlu-HWSYkLRPG-NH2 (pGlu : Pyroglutamic acid, k : DLysine) [54
Bombesin/Bn receptors Prostate, breast, pancreas and small cell lung cancer cell line yQWAV-βAla-HF-Nle-NH2 (y : DTyrosine, Nle : Norleucine, βAla : β-Alanine) [54,56
Vasoactive intestinal peptide receptors (VIP receptors) Breast, colon and endocrine cancer tumor cells HSDAVFTDNYTRLRKQMAVKKYLNSILN-NH2 [54
Neurotensin receptor 1 (NTSR1) Breast, colon and pancreatic cancer cells RRPYIL [54,57
CCK2R Liver, thyroid and pancreatic cancer cells eAYGWMDF-NH2 (e : DGlutamic acid) [54
MC1Melanoma cells Ac-Nle-DHfRWGK-NH2 (Ac : Acetyl, f : DPhenylalanine) [54,58
Human Y1 receptor (hY1R) Ewing sarcoma and breast cancer cell lines YPSKPDFPGEDAPAEDLARYYSALRHYINLITRPRY-NH2 [54
N-cadherin HUVEC SWTLYTPSGQSK [42
Carbonic anhydrase IX Renal cell carcinoma cell lines YNTNHVPLSPKY [42,59

Conjugation of ligands with the nanoparticles is mediated via covalent or non-covalent chemical conjugation. However, non-covalent conjugation often leads to weak bonding making it less efficacious. Covalent coupling is commonly achieved with the conjugations of the following groups (1) hydrazide-aldehyde, (2) carboxylic acid-primary amine, (3) thiol-thiol, (4) gold-thiol, (5) maleimide and thiol and (6) azide-alkyne. Different types of linkages formed due to various chemical conjugation reactions are described in Figure 3 [60,61].

Different types of linkages formed due to various chemical conjugation reactions for active targeting

Figure 3
Different types of linkages formed due to various chemical conjugation reactions for active targeting

Stability of the linkages under physiological conditions (pH 7.4): Hydrazide-aldehyde conjugation (acid-labile), amide bond (stable), thiol-thiol conjugation (cleaved under reducing condition), gold-thiol conjugation (stable), thiol/maleimide conjugation (stable), azide/alkyne conjugation (stable) and streptavidin-biotin conjugation (stable).

Figure 3
Different types of linkages formed due to various chemical conjugation reactions for active targeting

Stability of the linkages under physiological conditions (pH 7.4): Hydrazide-aldehyde conjugation (acid-labile), amide bond (stable), thiol-thiol conjugation (cleaved under reducing condition), gold-thiol conjugation (stable), thiol/maleimide conjugation (stable), azide/alkyne conjugation (stable) and streptavidin-biotin conjugation (stable).

Close modal

Size, shape and surface modification also remain essential to achieve effective tumor targeting (Figure 4). In order to specifically target the tumor, nanoparticles must first travel in circulation without being engulfed by macrophages [62]. Nanoparticles having a diameter of ∼5 nm undergo rapid renal clearance, whereas nanoparticles of 50–100 nm diameter mostly accumulate in the liver. Larger particles >2000 nm in diameter tend to accumulate in the spleen. It has been found that nanoparticles having a diameter of 100–200 nm can escape filtration by the liver and spleen (Figure 5) [10]. Researchers observed that nanoparticles of size ranges 30-50 nm diameter showed maximum cellular internalization ability [63]. Studies have also shown that the size of nanoparticles is also dependent on tumor maturity. Reports suggest that with increasing particle size, the area of permeation within tumors become smaller [64]. Gold nanoparticles with 15–45 nm diameter have been found to accumulate in tumors of 0.5-1 cm3 volume or above [65]. Recent findings suggest that the shape of the nanoparticles also determines the cellular uptake [66]. Several nanostructures like 2D polygonal shape, 3D polygonal shape, rod, snowflakes, flowers, thorns, hemispheres, cones, filaments, etc., have been designed to study their efficacies [62]. Recent studies have revealed that oblate shape is favoured in circulation [62]. Mitragotri and co-workers have modified a solid polystyrene microparticle into a red blood cell (RBC) shaped particle using layer-by-layer (LbL) self-assembly technique. The nanoparticles were synthesized using PLGA, which can be used as a carrier for drug and imaging agents [67]. Additionally, RBC-derived cell membrane and a hybrid membrane having membrane from RBC and cancer cell line were used for delivering chemotherapeutic drugs and such methodologies can be explored for designing personalized nanomedicine [68,69]. Surface charge of the nanoparticles also plays a significant role in nanoparticle internalization (Figure 4). Current studies have demonstrated that nanoparticles having a size range of 50–100 nm carrying a very slight positive charge favour the penetration in large tumors [11].

Structural components of multifunctional cancer nanomedicine

Figure 4
Structural components of multifunctional cancer nanomedicine

Commonly used therapeutic agents are chemotherapeutic drugs, RNAi therapeutics, and imaging agents include MRI contrast agents, radionuclides, fluorescent probes, etc. Therapeutic agents can either be covalently conjugated or non-covalently encapsulated. Attaching imaging probes with nanoparticles containing therapeutic agents make it a theranostic platform (this image was drawn based on the information provided in Chou et al. 2011 [70], Figure 1).

Figure 4
Structural components of multifunctional cancer nanomedicine

Commonly used therapeutic agents are chemotherapeutic drugs, RNAi therapeutics, and imaging agents include MRI contrast agents, radionuclides, fluorescent probes, etc. Therapeutic agents can either be covalently conjugated or non-covalently encapsulated. Attaching imaging probes with nanoparticles containing therapeutic agents make it a theranostic platform (this image was drawn based on the information provided in Chou et al. 2011 [70], Figure 1).

Close modal

Biodistribution of nanoparticles

Figure 5
Biodistribution of nanoparticles

Nanoparticles having different (A) size, (B) shape, (C) surface charge and their biodistribution in different organs. This figure will guide us for designing organ-specific delivery of nanoparticles (this image was drawn based on the information provided in Blanco et al. 2015 [10], Figure 5).

Figure 5
Biodistribution of nanoparticles

Nanoparticles having different (A) size, (B) shape, (C) surface charge and their biodistribution in different organs. This figure will guide us for designing organ-specific delivery of nanoparticles (this image was drawn based on the information provided in Blanco et al. 2015 [10], Figure 5).

Close modal

A transient increase in blood pressure during systemic administration also causes increased tumor-specific nanoscale drug delivery. For in vivo delivery, near the wall margination is favored that interacts with the tumor vasculature bed. RBCs tend to travel in the middle of the blood flow, creating a “cell-free layer”. Spherical nanoparticles follow the bloodstream whereas, rod-shaped nanoparticles undergo a lateral drift due to variable drag forces and torques. Nanorods show a 7-fold higher accumulation at the vessel lining than nanospheres. Again, discs marginate two times higher than rods. Particles are deposited at a higher rate at the site of the vessel bifurcation [62]. Additionally, the “multivalency” of a nanoparticle can be harnessed to enhance the binding affinity or avidity of ligand to receptor interaction [7]. It has been found that oblong-shaped nanoparticles are more helpful in forming more multivalent interactions compared to spherical nanoparticles (Figure 6) [62]. The cellular internalization process also depends on another factor termed “membrane wrapping time” (Figure 7). Smaller nanoparticles tend to dissociate faster from receptors before being engulfed by the membrane to achieve receptor-mediated endocytosis. Again, extremely large nanoparticles limit the process of membrane wrapping. Chan and co-workers suggested that 40–50 nm nanoparticles remain the critical cut off point for receptor mediated endocytosis [71].

Multivalent interaction of nanoparticles (having different shapes) with cell surface receptors

Figure 6
Multivalent interaction of nanoparticles (having different shapes) with cell surface receptors

(A) Role of the shape of nanoparticles on multivalent interaction with cell surface receptors. Compared with nanospheres, oblong-shaped nanoparticles can form more multivalent interactions, which is required for vascular targeting (this image was drawn on the basis of information provided in Blanco et al., 2015 [10], Figure 3). (B) Role of contact angle of nanoparticles in intracellular internalization. Rod-shaped nanoparticles tend to internalize faster when it is present perpendicularly on the cell membrane. Due to the symmetry of the spherical nanoparticles, they do not prefer any specific contact angle (This image was drawn on the basis of information provided in Toy et al., 2014 [62], Figure 2).

Figure 6
Multivalent interaction of nanoparticles (having different shapes) with cell surface receptors

(A) Role of the shape of nanoparticles on multivalent interaction with cell surface receptors. Compared with nanospheres, oblong-shaped nanoparticles can form more multivalent interactions, which is required for vascular targeting (this image was drawn on the basis of information provided in Blanco et al., 2015 [10], Figure 3). (B) Role of contact angle of nanoparticles in intracellular internalization. Rod-shaped nanoparticles tend to internalize faster when it is present perpendicularly on the cell membrane. Due to the symmetry of the spherical nanoparticles, they do not prefer any specific contact angle (This image was drawn on the basis of information provided in Toy et al., 2014 [62], Figure 2).

Close modal

Illustration of size (diameter) dependent nanostructure internalization due to membrane wrapping

Figure 7
Illustration of size (diameter) dependent nanostructure internalization due to membrane wrapping

This figure indicates that 40–50 nm gold nanostructures show optimum cellular uptake activity due to membrane wrapping. Smaller nanoparticles readily dissociate from receptors before being engulfed by the membrane, whereas extremely large nanoparticles fail to cause membrane wrapping (this figure was prepared based on the information provided in Jiang et al., 2008 [71], Figure 3A).

Figure 7
Illustration of size (diameter) dependent nanostructure internalization due to membrane wrapping

This figure indicates that 40–50 nm gold nanostructures show optimum cellular uptake activity due to membrane wrapping. Smaller nanoparticles readily dissociate from receptors before being engulfed by the membrane, whereas extremely large nanoparticles fail to cause membrane wrapping (this figure was prepared based on the information provided in Jiang et al., 2008 [71], Figure 3A).

Close modal

An ideal drug delivery platform should possess the ability to target and control the drug release, which facilitates sustained release of drugs [72]. Drug delivery often renders toxicity and side effects. These hurdles can be overcome by controlled drug release. This feature also leads to a high therapeutic index for the conjugated drug molecule. The binding of the drugs to the nanoparticles is achieved by adsorption, absorption, entrapment (the process of incorporation of a drug into a matrix) and covalent binding. The release of the drug molecules is decided on the basis of their solubility, desorption, diffusion through nanoparticles matrix, degradation of nanoparticle matrix and combination of such phenomena [73,74]. In many of such formulations, a phenomenon called “burst release” is often observed. A large bolus of drug is immediately released before it reaches to a stable profile. Researchers found that low molecular weight drugs are prone to exhibit burst release profile. Burst release may often lead to local or systemic toxicity, short half-life of drugs in vivo and shortened release profile [72]. Researchers have often modified the nanoparticles to get over this difficulty. Le and co-workers have used chitosan to modify PLGA nanoparticles carrying paclitaxel and monitored the drug release profile. They found that modification of PLGA nanoparticles with chitosan led to reduced burst release of drug [75]. Again, designed nanoparticles that are prone to transcytosis (the vesicular transport of macromolecules from one side of a cell to the other) possess the potential to reach tumor cells and show efficient anticancer efficacy [76].

Nanotechnology has made advancements in mediating anticancer therapy and imaging at the tumor microenvironment. Several synthetic and natural nanoparticles have been used currently. These include polymeric conjugates and polymeric nanoparticles ((N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers, Poly(lactic co-glycolic acid) (PLGA) copolymers, etc.; liposomes and micelles; synthetic organic nanoparticles such as dendrimers; carbon-based nanostructures such as carbon nanotubes (CNTs) and polyhydroxylated/ PEGylated fullerenes; inorganic nanoparticles of gold, silver, and iron oxide; quantum dots (QDs); viral capsids and ferritin, etc [7]. Liposomes are spherical vesicles comprising of one or more lipid bilayers, especially phospholipids. Liposomes have been widely used as a drug delivery vehicle. These increase the drug’s efficacy and therapeutic index by protecting the drug from the external environment. Dendrimers are radially symmetric branched polymeric nanoparticles. They possess the properties like poly valency, self-assembly, chemical stability, low toxicity and solubility. Dendrimers have been widely utilized to deliver anticancer drugs. Polymeric nanoparticles contain random or block co-polymers. These are colloidal particles having a size range of 1 to 1000 nm. Drug delivery has been widely performed using polymeric nanoparticles, either by encapsulation or chemical conjugation of drugs [18].

The delivery of chemotherapeutic drugs like cisplatin has been rigorously manipulated with nanotherapy to overcome their toxicity. Sengupta and co-workers have designed a novel cisplatin nanoparticle by harnessing PEG-functionalized poly-isobutylene-maleic acid (PEG-PIMA) co-polymer. Cisplatin is released in a pH-dependent manner and shows improved antitumor efficacy both in vitro and in vivo with limited nephrotoxicity [77]. They have also engineered another novel polymer glucosamine-functionalized PIMA to complex with platinum at a unique platinum to polymer ratio. Such nanoparticles also exhibit improved efficacy against breast and lung cancer with reduced systemic and nephrotoxicity [78]. Sengupta et al. have also designed cholesterol-tethered platinum II-based supramolecular nanoparticle with increased efficacy and reduced toxicity [79]. Prolonged use of cisplatin leads to nephrotoxicity. In order to overcome this limitation, other next-generation platinum-based drugs have been developed. Carboplatin (cis-diamino-(1,1-cyclobutandicarboxylate)platinum(II)) has been used recently, but it shows a cross-resistance with cisplatin. Oxaliplatin (cis-[(1R,2R)-1,2-cyclohexanediamine-N,N′]-oxalatoplatinum (II)) does not show such cross-resistance with cisplatin and is also highly soluble in water. Moreover, trans-1,2-diaminocyclohexane (DACH) ring of oxaliplatin adduct fills the major groove of DNA more efficiently than cisplatin. Scientists have derivatized the monomeric units of a PIMA copolymer with glucosamine, which chelates DACH platinum (II) and releases DACH-platinum in a sustained pH-dependent manner with reduced systemic toxicity and minimal kidney accumulation [80]. Kulkarni et al. also designed a computational algorithm to develop nanoscale supramolecular structures for cancer treatment [81].

Nanotechnologies have also been currently used for genetic treatments by nanoparticle-mediated delivery of RNAi therapeutics [10]. The primary challenge of RNAi-based therapeutics for its successful translation to clinics is the instability of RNA molecules, their rapid degradation in presence of nuclease and poor cellular uptake because of its highly anionic nature [82]. Genetic material, such as antisense oligonucleotides, mRNAs and siRNAs, and in the specific case of plasmid DNA have been used to achieve gene therapy via nanoparticles.

Peptides have been used for targeted delivery of diagnostics and chemotherapeutic agents for anticancer therapy. Peptides remain advantageous over other nanotherapeutics in terms of their self-assembling property, easy synthesis, structural manipulation to achieve protease stability, functionalization property, conjugation to the cell surface receptors and maximum therapeutic efficacy of cargo. Peptides show minimal toxicity, improved biodegradability, rapid renal clearance, and remain stable at physiological conditions. Anticancer agents like paclitaxel/docetaxel, doxorubicin, curcumin, fluorouracil have been successfully loaded on to self-assembled peptides and evaluated for their preclinical and clinical status [83]. Peptides have also been used as a potential molecular transporter based nanosystem to deliver RNAi therapeutics like siRNA [84–86].

Table 4 enlists cancer therapeutics that either have received FDA approval or are currently undergoing clinical evaluation [87].

Table 4
Nanomaterials in clinical use
NanomaterialTrade nameCompositionApplicationManufacturerCurrent statusAdverse effects
Metallic 
Iron oxide NanoTherm Iron oxide NP conjugate with surface ligand aminosilane Prostate cancer MagForce Phase 2b clinical trial Acute urinary retention 
 Feraheme®; Iron oxide nanoparticles (coated with polyglucose sorbitol carboxymethylether). Imaging agent for triple-negative breast cancer, head and neck cancer, nonsmall cell lung cancer etc. AMAG Pharmaceuticals, Inc. Phase 3 clinical trial Constipation, fluid retention in the legs, feet, arms or hands, headache, nausea 
Gold Aurimmune Tumor necrosis factor (TNF)-gold nanoparticle Cancer therapy (various cancer types) CytImmune Sciences Phase 1 clinical trial Fever 
 Aurolase Silica-gold nanoshells coated with PEG Thermal ablation of solid tumors: head/neck cancer, primary and/or metastatic lung tumors Nanospectra Pilot study Inflammation 
Nanoshells Auroshell Gold metal shell and a non-conducting silica core Cancer therapy (head and neck) Nanospectra Biosciences Phase 1 clinical trial Under investigation 
Organic 
Protein Abraxane Albumin-bound Paclitaxel for Injectable Suspension Cancer therapy (breast) Abraxis Bioscience FDA approved Cytopenia 
Liposome Doxil/Caelyx Liposomal Doxorubicin Cancer therapy Ortho Biotech FDA approved Hand-foot syndrome, stomatitis 
Polymer Oncaspar Pegylated form of L-asparaginase Cancer therapy (acute lymphoblastic leukemia) Rhône-Poulenc Rorer Phase 2 clinical trial Urticaria, rash 
 CALAA-01 Formulation of siRNA that consists of a CD-polycation, adamantane (AD)–PEG (MW of 5000) conjugate and AD-PEG-transferrin as the targeting ligand, Cancer therapy (various cancer types) Calando Phase 2 clinical trial Mild renal toxicity 
Micelle Genexol-PM Paclitaxel-methoxy polyethylene glycols (mPEG)-Poly(D,L-Lactic Acid) (PDLLA) conjugate Cancer therapy (Various cancer types) Samyang Phase 4 clinical trial Peripheral sensory neuropathy, neutropenia 
NanomaterialTrade nameCompositionApplicationManufacturerCurrent statusAdverse effects
Metallic 
Iron oxide NanoTherm Iron oxide NP conjugate with surface ligand aminosilane Prostate cancer MagForce Phase 2b clinical trial Acute urinary retention 
 Feraheme®; Iron oxide nanoparticles (coated with polyglucose sorbitol carboxymethylether). Imaging agent for triple-negative breast cancer, head and neck cancer, nonsmall cell lung cancer etc. AMAG Pharmaceuticals, Inc. Phase 3 clinical trial Constipation, fluid retention in the legs, feet, arms or hands, headache, nausea 
Gold Aurimmune Tumor necrosis factor (TNF)-gold nanoparticle Cancer therapy (various cancer types) CytImmune Sciences Phase 1 clinical trial Fever 
 Aurolase Silica-gold nanoshells coated with PEG Thermal ablation of solid tumors: head/neck cancer, primary and/or metastatic lung tumors Nanospectra Pilot study Inflammation 
Nanoshells Auroshell Gold metal shell and a non-conducting silica core Cancer therapy (head and neck) Nanospectra Biosciences Phase 1 clinical trial Under investigation 
Organic 
Protein Abraxane Albumin-bound Paclitaxel for Injectable Suspension Cancer therapy (breast) Abraxis Bioscience FDA approved Cytopenia 
Liposome Doxil/Caelyx Liposomal Doxorubicin Cancer therapy Ortho Biotech FDA approved Hand-foot syndrome, stomatitis 
Polymer Oncaspar Pegylated form of L-asparaginase Cancer therapy (acute lymphoblastic leukemia) Rhône-Poulenc Rorer Phase 2 clinical trial Urticaria, rash 
 CALAA-01 Formulation of siRNA that consists of a CD-polycation, adamantane (AD)–PEG (MW of 5000) conjugate and AD-PEG-transferrin as the targeting ligand, Cancer therapy (various cancer types) Calando Phase 2 clinical trial Mild renal toxicity 
Micelle Genexol-PM Paclitaxel-methoxy polyethylene glycols (mPEG)-Poly(D,L-Lactic Acid) (PDLLA) conjugate Cancer therapy (Various cancer types) Samyang Phase 4 clinical trial Peripheral sensory neuropathy, neutropenia 

Signal transduction pathways play a crucial role in cellular functions like survival, growth, differentiation and metabolism. In the case of cancer, such signaling pathways remain altered, leading to uncontrolled proliferation, immortality and tumorigenesis. Researchers have focused on identifying the drug targets to inhibit oncogenic signaling pathways [28]. Classically, humanized antibodies and small molecule inhibitors have been used as potential inhibitors to target such oncogenic pathways. But nanotherapy has helped target cancer cells selectively without causing toxicity to the healthy cells. The following section mentions key signaling pathways responsible for tumorigenesis and the approaches taken to target such pathways.

Receptor tyrosine kinase (RTK) plays a crucial role in major cellular processes like proliferation. Growth factors bind to RTKs, resulting in the dimerization and activation of RTKs. The activation of the intracellular kinase domain of RTKs triggers downstream pathways (Figure 8) [28]. Different types of RTKs have been identified depending on growth factor ligands (e.g., epidermal growth factor receptor [EGFR], vascular endothelial growth factor receptor [VEGFR], fibroblast growth factor receptors [FGFR], platelet-derived growth factor receptor [PDGFR] etc.). In cancer, RTKs are aberrantly activated and their mutations lead to various disorders. Therefore, RTKs and their ligands remain a potential drug target [28]. The inhibitors are mentioned in Figure 8.

Oncogenic receptor tyrosine kinase pathway with its downstream signaling pathways and small molecule inhibitors targeting different proteins of the network

Figure 8
Oncogenic receptor tyrosine kinase pathway with its downstream signaling pathways and small molecule inhibitors targeting different proteins of the network

Receptor tyrosine kinase (RTK) plays a significant role in cellular proliferation. MAPK and PI3K-AKT-mTOR pathways are two key downstream pathways of RTK. Targeting these pathways has remained a successful approach to cause the antitumor effect. Small molecule inhibitors have been widely used for targeting different components of such pathways. Several small molecule inhibitors targeting different proteins of RTK signaling pathways are depicted in this diagram.

Figure 8
Oncogenic receptor tyrosine kinase pathway with its downstream signaling pathways and small molecule inhibitors targeting different proteins of the network

Receptor tyrosine kinase (RTK) plays a significant role in cellular proliferation. MAPK and PI3K-AKT-mTOR pathways are two key downstream pathways of RTK. Targeting these pathways has remained a successful approach to cause the antitumor effect. Small molecule inhibitors have been widely used for targeting different components of such pathways. Several small molecule inhibitors targeting different proteins of RTK signaling pathways are depicted in this diagram.

Close modal

Kulkarni et al. found that multi-receptor tyrosine kinase inhibitor (XL184) loaded liposomes mediated nanoscale medicine improves intratumoral concentration, enhances antitumor efficacy and reduces toxicities [88].

Targeting the downstream signaling pathways also act as an alternative strategy. Mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase PI3K-AKT-mTOR (mammalian target of rapamycin) pathways are two critical downstream pathways [28].

MAPK pathway comprises a series of proteins like rat sarcoma virus (RAS), rapidly accelerated fibrosarcoma (RAF), mitogen-activated protein kinase kinase (MEK) and extracellular signal-regulated kinase 1/2 (ERK 1/2). This pathway is up-regulated in most cancer types and responsible for abnormal proliferation leading to tumorigenesis. RAS, RAF and MEK gained the most attention as therapeutic targets [28]. The progress in the therapeutic strategy targeting MAPK pathway using small molecule inhibitors is mentioned in Figure 8.

Researchers found that PD98059, a selective MAPK inhibitor, conjugated with hexadentate-poly-D,L-lactic acid-co-glycolic acid polymer potentiate the anticancer efficacy of cisplatin chemotherapy [89].

Phosphoinositide 3-kinase (PI3K) pathway is responsible for cellular processes like proliferation, growth, survival and apoptosis. This pathway is mutated in 30% of all human tumors. Activated RTKs set off several downstream signaling cascades, especially protein kinase B (Akt) and mammalian target of rapamycin (mTOR), responsible for various cellular functions [28]. The small molecule inhibitors targeting this oncogenic signaling pathway are mentioned in Figure 8.

Utilizing the nanotechnology, Harfouche et al. found that encapsulating LY294002, a PI3 kinase inhibitor into biodegradable PLGA nanoparticle causes successful inhibition of Akt phosphorylation resulting in the inhibition of proliferation [90]. Sengupta and co-workers have rationally conjugated PI3K inhibitors (PI103 and PI828) using a cholesterol-based derivative, enabling supramolecular nano assembly with L-α-phosphatidylcholine and DSPE-PEG [1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polythylene glycol)] to achieve increased antitumor efficacy [91]. They have also suggested that a rational combination of cis-platinum nanoparticles and a PI3K-targeted therapeutic remains a potential therapy for breast cancer [92]. Free nanoparticles or nanoparticles carrying different therapeutic agents like oligonucleotides, cytotoxic drugs, antibodies, etc., have been used to target different oncogenic pathways (Table 5).

Table 5
Nanoparticles used to target signaling pathways
NanocarriersMaterials/drugsCell line/animal modelEffectsRef.
Gelatin nanoparticles (D-NPs: gelatin nanoparticles loaded with NF-kB inhibiting decoy oligodeoxynucleotides) NF-kB inhibiting decoy oligodeoxynucleotides
(Decoy oligonucleotides (decoy ONs) epitomize an ideal pharmacological tool to selectively block NF-κB activation.) 
Kupffer cells Inhibition of NF-κB activation by D-NPs in kupffer cells (KC) D-NPs inhibit the nuclear translocation of p65, a common subunit of NF-kB. Improve survival and reduction in liver damage [93
Folate-linked lipid-based nanoparticles NF-kB decoy RAW264.7 The NF-κB decoy shows an inhibitory effect in cytoplasm (inhibition of NF-kB translocation into nucleus of LPS-activated macrophages)
Inhibit particular matter (size ≤2.5µm) (PM2.5) induced neuroinflammation 
[94
Fisetin nanoparticles (FN)
(Fisetin, a natural flavonoid) 
No cargo C57BL/6 mice primary astrocytes Restrict PM2.5 exposure-induced NF-κB signaling activation.
Decrease PM2.5-induced astrocytes activation.
Reduce pro-inflammatory cytokines IL-1β and TNF-α 
[95
mPEG-PLGA nanoparticles Benzoylaconitine RBCs, RAW264.7 cells Inhibit the expression of NF-κB p65
Inhibition of NF-κB signaling to reduce inflammation 
[96
Silica nanoparticles (MSN-mesoporous silica nanoparticles) NF-κB p65 antibody (p65, also known as RelA) Balb/c mice Translocation and cell signaling transduction (the nanoparticle binds to p65, forming a complex, thereby inhibiting the entry of p65 into nucleus) [97
Nano-Selenium (Nano-Se) No cargo Albino wistar rats Exhibit negative NF-κB immune expression.
Reduces pancreatic injury and improves pancreatic functions 
[98
Astragalus polysaccharide nanoparticles
(Astragalus polysaccharide (APS) is a water-soluble heteropolysaccharide with bioactive effects, A. Membranaceus stems or dried roots derivative) 
No cargo H9c2 cells/ C57BL/6 mice Inhibition of TLR4/ NF-κB pathway activation
Decrease the secretion of proinflammatory cytokine 
[99
Niosome nanoparticle (vesicles composed of biodegradable non-ionic surfactants, which is an alternative to liposomes) Curcumin Human glioblastoma stem-like cells (GSCs) Reduce the mRNA expressions of NF-κB and IL-6 and increase the expression of Bcl2
Induce cell cycle arrest, ROS generation and apoptosis
Anti-tumor effect 
[100
ABI-009 (albumin-bound-rapamycin nanoparticle) Rapamycin Gastroenteropancreatic or lung neuroendocrine tumors patients mTOR
pathway inhibitor targeting cells with TSC1/TSC2 genes mutations 
[101
Mesoporous silica nanoparticles γ-secretase inhibitors (GSIs) FVB/N adult mice MSNPs carrying GSIs used block Notch signaling [102
Gold nanoparticles (AuNPs) No cargo C3H/HeN mice, NOD-SCID mice AuNPs could deactivate TGF- β1(cys-rich protein) by directly binding to the cysteine buried inside the protein through covalent bonds (S–Au bonds ≈ 40 kcal mol-1), disrupting the disulfide bond in the protein, thus destroying the structure and unfolding the protein. [103
Anthothecol-encapsulated PLGA-nanoparticles (Antho-NPs) Anthothecol (Anthothecol, is a limonoid isolated from a plant named Khaya anthotheca (Meliaceae), which acts as an antimalarial compound) AsPC-1,PANC-1 and Mia-Paca-2 cell Antho-NPs are found to actively inhibit the expression of Gli, Patched1, Bcl-2 and CyclinD1 in pancreatic CSCs.
Antho-NPs suppresses cell motility, migration and invasion by up-regulating E-cadherin and inhibiting N-cadherin and Zeb1 
[104
NanocarriersMaterials/drugsCell line/animal modelEffectsRef.
Gelatin nanoparticles (D-NPs: gelatin nanoparticles loaded with NF-kB inhibiting decoy oligodeoxynucleotides) NF-kB inhibiting decoy oligodeoxynucleotides
(Decoy oligonucleotides (decoy ONs) epitomize an ideal pharmacological tool to selectively block NF-κB activation.) 
Kupffer cells Inhibition of NF-κB activation by D-NPs in kupffer cells (KC) D-NPs inhibit the nuclear translocation of p65, a common subunit of NF-kB. Improve survival and reduction in liver damage [93
Folate-linked lipid-based nanoparticles NF-kB decoy RAW264.7 The NF-κB decoy shows an inhibitory effect in cytoplasm (inhibition of NF-kB translocation into nucleus of LPS-activated macrophages)
Inhibit particular matter (size ≤2.5µm) (PM2.5) induced neuroinflammation 
[94
Fisetin nanoparticles (FN)
(Fisetin, a natural flavonoid) 
No cargo C57BL/6 mice primary astrocytes Restrict PM2.5 exposure-induced NF-κB signaling activation.
Decrease PM2.5-induced astrocytes activation.
Reduce pro-inflammatory cytokines IL-1β and TNF-α 
[95
mPEG-PLGA nanoparticles Benzoylaconitine RBCs, RAW264.7 cells Inhibit the expression of NF-κB p65
Inhibition of NF-κB signaling to reduce inflammation 
[96
Silica nanoparticles (MSN-mesoporous silica nanoparticles) NF-κB p65 antibody (p65, also known as RelA) Balb/c mice Translocation and cell signaling transduction (the nanoparticle binds to p65, forming a complex, thereby inhibiting the entry of p65 into nucleus) [97
Nano-Selenium (Nano-Se) No cargo Albino wistar rats Exhibit negative NF-κB immune expression.
Reduces pancreatic injury and improves pancreatic functions 
[98
Astragalus polysaccharide nanoparticles
(Astragalus polysaccharide (APS) is a water-soluble heteropolysaccharide with bioactive effects, A. Membranaceus stems or dried roots derivative) 
No cargo H9c2 cells/ C57BL/6 mice Inhibition of TLR4/ NF-κB pathway activation
Decrease the secretion of proinflammatory cytokine 
[99
Niosome nanoparticle (vesicles composed of biodegradable non-ionic surfactants, which is an alternative to liposomes) Curcumin Human glioblastoma stem-like cells (GSCs) Reduce the mRNA expressions of NF-κB and IL-6 and increase the expression of Bcl2
Induce cell cycle arrest, ROS generation and apoptosis
Anti-tumor effect 
[100
ABI-009 (albumin-bound-rapamycin nanoparticle) Rapamycin Gastroenteropancreatic or lung neuroendocrine tumors patients mTOR
pathway inhibitor targeting cells with TSC1/TSC2 genes mutations 
[101
Mesoporous silica nanoparticles γ-secretase inhibitors (GSIs) FVB/N adult mice MSNPs carrying GSIs used block Notch signaling [102
Gold nanoparticles (AuNPs) No cargo C3H/HeN mice, NOD-SCID mice AuNPs could deactivate TGF- β1(cys-rich protein) by directly binding to the cysteine buried inside the protein through covalent bonds (S–Au bonds ≈ 40 kcal mol-1), disrupting the disulfide bond in the protein, thus destroying the structure and unfolding the protein. [103
Anthothecol-encapsulated PLGA-nanoparticles (Antho-NPs) Anthothecol (Anthothecol, is a limonoid isolated from a plant named Khaya anthotheca (Meliaceae), which acts as an antimalarial compound) AsPC-1,PANC-1 and Mia-Paca-2 cell Antho-NPs are found to actively inhibit the expression of Gli, Patched1, Bcl-2 and CyclinD1 in pancreatic CSCs.
Antho-NPs suppresses cell motility, migration and invasion by up-regulating E-cadherin and inhibiting N-cadherin and Zeb1 
[104

Another approach to target oncogenic signaling pathways is mediated by siRNA delivery, a component of gene therapy. Peptides have also been used as a molecular transporter-based nano-system to deliver siRNA targeting signaling pathways [86,105,106,107]. This is discussed in the later section of this review.

Angiogenesis is a phenomenon of the formation of the new blood capillaries out of pre-existing blood vessels. This phenomenon is essential for wound healing [108–111]. Cancer metastasis is also positively correlated with angiogenesis [112]. Therefore, blocking angiogenesis is a practical approach to inhibit cancer progression. Commonly available anti-angiogenic therapy harbors any of these two strategies: (1) damaging the existing blood vessels or (2) preventing the formation of new blood capillaries [113]. Anti-angiogenic therapy is different from commonly used tumor-targeted chemotherapy. It selectively targets the tumor-associated vasculature instead of the tumor cells, shows increased bioavailability for systemically administered drugs compared with tumor-targeted therapy and requires a low administration dose, leading to lesser systemic toxicity [113,114].

In spite of these advantages, chemotherapy-mediated anti-angiogenic therapy suffers from certain limitations. The tumor vasculature remains unreachable to most of the anti-angiogenic inhibitors. Nanotechnology has become an advanced and effective method to address this problem. Nanoparticle-mediated delivery of therapeutics has been achieved following both passive and active targeting [113]. Researchers have shown that conjugating cytotoxic drugs or angiogenic inhibitors with nanoparticles prefer to home at the tumor site following the EPR effect [115,116]. Active targeting has also been effective in targeting angiogenesis. Anti-angiogenic therapy harbors targeting VEGFR, αvβ3 integrins and other angiogenic factors. Synthetic peptides having the recognition site for integrins (cyclic Arginine-Glycine-Aspartic acid [cRGD]) have been widely used for targeted nanotherapy [113]. A plethora of nano-vectors have been reported by researchers for nanotechnology-based anti-angiogenic therapy, including polymeric nanoparticles, liposomes and micelles, dendrimers, carbon nanostructures, inorganic nanoparticles (e.g., gold, silver and iron oxide), etc. Recent nanotechnology-based anti-angiogenic therapies also use a gene silencing-based approach using therapeutic genes or siRNA [113].

A few reports on nanotechnology-based anti-angiogenic therapy are provided in Table 6.

Table 6
Nanoparticles used for anti-angiogenic therapy
NanoparticleCargoUsed againstCommentsRef.
Polymer 
HPMA TNP-470 (Caplostatin) Human melanoma, lung carcinoma Prevents crossing of blood–brain barrier (BBB), limiting neurotoxicity [117
 Aminobisphosphonate drug alendronate (Fosamax) and paclitaxel/TNP-470 Osteosarcoma Inhibits bone metastasis [118
 Radionuclidelabeled, cyclized RGD peptide Solid tumors Used for diagnosis and therapeutic application [119
PLGA LY294002 (PI3K pathway inhibitor) Zebrafish melanoma, breast adenocarcinoma Shows anti-angiogenic effect [90
PLGA nanoparticle encapsulated within PEG linked lipid envelop Doxorubicin (covalently attached to inner PLGA core) and anti-angiogenic agent (combretastatin) Melanoma Termed as “nanocell,” shows an improved therapeutic index with reduced toxicity [115
PEGylated polyethyleneimine (PEI) consisting RGD peptide PEGylated polyethyleneimine (PEI) consisting RGD peptide sFlt-1 gene Colon carcinoma Blocks VEGF binding to membrane-bound Flt-1 receptor and inhibits proliferation [120
 VEGFR-2 targeting siRNA Mouse neuroblastoma Inhibits angiogenesis [121
Polysaccharides and dendrimers 
Chitosan coated poly-isohexylcyanoacrylate nanoparticle Anti-RhoA siRNA Breast cancer mouse xenograft model Inhibits tumor growth and metastasis [122
Boronated polyamidoamine dendrimer VEGF121 Colon carcinoma in mice Shows anti-angiogenic effect [123
Lipid-based nanoparticles (liposome and micelle) 
Monomethoxy-polyethyleneglycolpolylactic acid copolymer TNP-470 Mouse melanoma Forms a micelle termed “Lodamine” Inhibits angiogenesis [124
Poly(ε-caprolactone)-polyethyleneglycol (PCL-PEG) Cyclic RGD pentapeptide (conjugated) and Doxorubicin (loaded) Kaposi’s sarcoma Forms a nanopolymeric micelle Shows antitumor activity [125
Ala-Pro-Arg-Pro-Gly (APRPG) peptide (for active targeting), PEG and hydrophobic anchor distearoylphosphatidylethanolamine (DSPE) Adriamycin Colon carcinoma Shows antiangiogenic effect [126
Neutral liposome Protein activated receptor-1 (PAR-1) siRNA Mouse melanoma Inhibits metastasis [127
Carbon nanostructures 
Fullerenols Doxorubicin Mouse melanoma Shows anti-angiogenic effect 119 
Inorganic nanoparticles 
Dextran coated iron oxide nanoparticles Radiolabeled anti-VEGF monoclonal antibody Liver cancer in mice Destruction of tumor with increased imaging resolution [128
Folate receptor targeted superparamagnetic iron oxide nanoparticle Doxorubicin Liver cancer Does not show systemic toxicity [129
PEGylated gold nanoparticle Doxorubicin Liver cancer in mice Shows antitumor activity [130
NanoparticleCargoUsed againstCommentsRef.
Polymer 
HPMA TNP-470 (Caplostatin) Human melanoma, lung carcinoma Prevents crossing of blood–brain barrier (BBB), limiting neurotoxicity [117
 Aminobisphosphonate drug alendronate (Fosamax) and paclitaxel/TNP-470 Osteosarcoma Inhibits bone metastasis [118
 Radionuclidelabeled, cyclized RGD peptide Solid tumors Used for diagnosis and therapeutic application [119
PLGA LY294002 (PI3K pathway inhibitor) Zebrafish melanoma, breast adenocarcinoma Shows anti-angiogenic effect [90
PLGA nanoparticle encapsulated within PEG linked lipid envelop Doxorubicin (covalently attached to inner PLGA core) and anti-angiogenic agent (combretastatin) Melanoma Termed as “nanocell,” shows an improved therapeutic index with reduced toxicity [115
PEGylated polyethyleneimine (PEI) consisting RGD peptide PEGylated polyethyleneimine (PEI) consisting RGD peptide sFlt-1 gene Colon carcinoma Blocks VEGF binding to membrane-bound Flt-1 receptor and inhibits proliferation [120
 VEGFR-2 targeting siRNA Mouse neuroblastoma Inhibits angiogenesis [121
Polysaccharides and dendrimers 
Chitosan coated poly-isohexylcyanoacrylate nanoparticle Anti-RhoA siRNA Breast cancer mouse xenograft model Inhibits tumor growth and metastasis [122
Boronated polyamidoamine dendrimer VEGF121 Colon carcinoma in mice Shows anti-angiogenic effect [123
Lipid-based nanoparticles (liposome and micelle) 
Monomethoxy-polyethyleneglycolpolylactic acid copolymer TNP-470 Mouse melanoma Forms a micelle termed “Lodamine” Inhibits angiogenesis [124
Poly(ε-caprolactone)-polyethyleneglycol (PCL-PEG) Cyclic RGD pentapeptide (conjugated) and Doxorubicin (loaded) Kaposi’s sarcoma Forms a nanopolymeric micelle Shows antitumor activity [125
Ala-Pro-Arg-Pro-Gly (APRPG) peptide (for active targeting), PEG and hydrophobic anchor distearoylphosphatidylethanolamine (DSPE) Adriamycin Colon carcinoma Shows antiangiogenic effect [126
Neutral liposome Protein activated receptor-1 (PAR-1) siRNA Mouse melanoma Inhibits metastasis [127
Carbon nanostructures 
Fullerenols Doxorubicin Mouse melanoma Shows anti-angiogenic effect 119 
Inorganic nanoparticles 
Dextran coated iron oxide nanoparticles Radiolabeled anti-VEGF monoclonal antibody Liver cancer in mice Destruction of tumor with increased imaging resolution [128
Folate receptor targeted superparamagnetic iron oxide nanoparticle Doxorubicin Liver cancer Does not show systemic toxicity [129
PEGylated gold nanoparticle Doxorubicin Liver cancer in mice Shows antitumor activity [130

Gene therapy is the modulation of gene expression towards treating a disease by cellular delivery of therapeutic nucleic acid. It holds unique promise in alteration of specific tumor genes functioning via gene addition, gene correction or gene knockdown [16]. Several approaches of cancer gene therapy include (1) suicide gene therapy: introduction of an enzyme expressing transgene into the cell that converts inactive prodrug into cytotoxic metabolite for host cells, (2) gene silencing: suppression of gene expression by RNAi techniques like siRNA, shRNA, antisense oligonucleotide, miRNA, etc., and (3) DNA vaccine: introduction of specific antigen encoding plasmid DNA into the cell to induce immune response.

Gene therapy shows the potential to deal undruggable targets to treat cancer as compared with conventional treatment by targeting cancer associated genes. Gene therapy can provide a solution to low bioavailability, reducing immune system based recognition and delivery of the gene regulators [131]. The fundamental challenge in the engineering of gene therapy is the development of clinically safe and effective delivery vectors. In clinics, both viral and non-viral mode of delivery is being used for systemic gene delivery [132]. The viral delivery systems are associated with various safety concerns as well as limited payload capacity and difficulty in large-scale production [16]. These factors led to the development of interest toward non-viral synthetic vectors for gene therapy. The non-viral vectors are advantageous in providing higher safety profile, low cost, large scale manufacturing potential, stability and higher payload [132]. Nanoparticle and nanoscale gene delivery vectors have emerged as efficient candidates for intracellular or systemic gene delivery.

Nanoparticle exploitation for gene delivery can be categorized into four groups : (1) lipid-based nanoparticles, (2) polymer-based nanoparticle, (3) peptide-based nanoscale material and (4) inorganic nanoparticles.

Lipid-based nanoparticles are the most widely used non-viral gene delivery vehicle [132,133]. Cationic liposomes are amphiphilic molecules which are made up of cationic polar head group, a hydrophobic domain and a linker connecting the polar head group with the non-polar tail. This cationic liposomes are routinely used for gene delivery [134]. Incorporation of longer lipidic chain (having around 18 methylene group, which can span the whole membrane) having unsaturation and small polar head facilitates the formation of conical shaped lipid in anionic membrane environment, which promotes the hexagonal phase transition of the lipid bilayer from the lamellar phase [135]. Hexagonal phase of the lipid bilayer is more fusogenic than lamellar phase. This transition of lamellar phase to hexagonal phase of lipid bilayer leads to cellular internalization and endosomal release of the internalized lipid-based nanoparticle [135]. Lipid nanoparticle containing ionizable lipids are designed in a way that it gets protonated at endosomal pH range (4.5–6.5). The protonated cationic lipid interacts with the anionic lipid of the endosomal membrane, leading to the transition of the lamellar phase to the hexagonal phase of lipid bilayer. Several liposomal nanoformulations have been in clinical development like DOTAP-cholesterol, GAP-DMORIE-DPyPE, etc. The FDA approval of Alnylam’s patisiran in 2018, is the first ever drug to successfully harness RNA interference to silence disease associated gene expression was a key milestone. Further, FDA approved lipid based nanoparticles like givosiran, lumasiran established a benchmark for lipid nanoparticle based drugs. Hou et al. have discussed in detail about the use of lipid based nanoparticles for gene delivery, their drawback and engineering principles for engineering next-generation improved lipid based nanoparticles [136].

The chemical diversity and functionalization potential of cationic polymer based nanoparticle makes it an attractive class of non-viral gene delivery vehicle. Polymers are considered as inert but certain modification by biologically active agents and counterions like spermidine or cell penetrating peptides improves the surface functionalization, nucleic acid loading and particle transfection [16] PLGA nanoparticle is capable of delivering nucleic acid with minimal toxicity but exhibit low transfection efficiency [16]. PLGA nanoparticles are surface functionalization with cell targeting or cell penetrating peptides to improve nucleic acid loading and cell penetrability. The condensation of negative phosphate bonds of nucleic acid with the cationic polymers into polyplexes protects nucleic acid from degradation during circulation [16].

Polyethylenimine (PEI) and its variants are among the most studied polymeric materials for gene delivery. The presence of nitrogen at every third position of the polymer increases the charge density and reduces the pH [132,137]. Various polymer-based nanoparticles such as dendrimers, polyion complex micelles (PICs), cyclodextrin, etc., have also been explored greatly in gene therapy. The polymeric nanoparticle system owing to their facile synthesis and flexible properties proves to be a new promising material for developing non-viral gene delivery system. The cationic polymer can be conjugated with the negatively charged genetic material via electrostatic attraction at physiological pH and facilitating gene delivery. Ekladious et al. have discussed in detail about the rational designing, physicochemical characteristics and advancements in different classes of polymer based delivery vehicle and their application in different fields including gene therapy [138].

Cell penetrating peptides (CPP) are a class of peptides facilitating the cellular internalization of nucleic acid based therapeutics either by covalent or non-covalent conjugation. The cellular uptake of the CPP peptide depends upon their sequence, structure, concentration and cell lines used for study [139]. The cellular internalization of cell penetrating peptides follow either by one or by the combination of the following mechanisms: (1) creating a transient pore in cell membrane, (2) endocytic uptake and (3) receptor mediated uptake. Depending upon the sequence, the CPP can be of cationic CPP or amphiphilic CPP.

The cationic CPPs are arginine or lysine rich short peptides. Arginine rich sequences are found to show enhanced cellular uptake. The arginine can form a more efficient bidentate hydrogen bond than the monodentate hydrogen bond formed by lysine with the anionic moieties like phosphate (PO43−), carbonate (CO32−) and sulfate (SO42−) on the cell surface [140]. These interactions facilitate the cellular internalization of the arginine or lysine residue enriched cationic peptides.

Amphiphilic peptides show enhanced cellular uptake by the formation of lipid rafts in the cell membrane. The cationic part of the CPP interacts with the negatively charged therapeutic nucleic acid and the hydrophobic part facilitates the cellular internalization of the peptide by interacting with the lipid bilayer [141,142].

N-Methylpurine DNA Glycosylase or MPG (GALFLGFLGAAGSTMGAWSQPKKKRKV) peptide is a great example of amphiphilic CPP. The hydrophobic stretch of this amphiphilic CPP (underlined) adopts a transient β-sheet structure creating a temporary channel in the cell membrane allowing the peptide–nucleic acid complex to internalize [143,144]. Amphiphilic α-helical peptide like penetratin (RQIKIWFQNRRMKWKK) generally remains unstructured in an aqueous solution but tends to adopt an α-helical conformation while interacting with cell membrane [84]. Majority of CPP internalizes via endosomal pathway and are accumulated inside endosome. Two strategies employed for endosomal escapes are use of (1) conformation changing fusogenic peptides and (2) proton buffering peptides [84]. The conformation changing fusogenic peptides in endosomal pH (4.5–6.5) undergoes a conformational change and ruptures the endosomal membrane by forming an amphiphilic helix [84]. These peptides are rich in histidine and glutamic acid and are sensitive to pH change. pH sensitivity arises due to imidazole group of histidine side chain having pKa value of ∼ 6.0 and glutamic acid side chain with pKa ∼ 4.3 allowing easy protonation and de-protonation of these amino acids in endosomal environment.

Proton buffering peptides escape the endosomal entrapment by accumulation of proton absorbing peptides inside endosome and disrupting the endosomal membrane by proton sponge effect. Imidazole ring of histidine having a pKa value of nearly 6.0 shows high buffering effect by acting as a weak base inside endosome. This causes influx of protons into the endosome and osmotic swelling causing endosomal rupture. Peptides provide advantage as a delivery vehicle due to their biocompatibility, biodegradability and sheer limitless combinations and modifications of amino acid residues inducing the assembly of modular, multiplexed systems [145]. Tarvirdipour et al. have discussed in detail the designing principles and attractive features of peptide based nanoscale materials for gene therapy [145].

This includes carbon nanotubes, magnetic nanoparticles, calcium phosphate nanoparticles, gold nanoparticles and quantum dots that are commonly used for gene delivery vehicles. The inorganic nanoparticles are resistant to microbial attacks and provide good storage stability [146]. The functionalized single-walled nanotubes are reported to enter premyelocytic leukemia and T-cell very easily, this ability is exploited to deliver nucleic acid into mammalian cells [147]. Water soluble allotrope of carbon (C60) fullerenes modified to aminofullerenes has positive charge on them. This aminofullerenes are reported to have high transfection efficacy of DNA into mammalian cells [148]. Inorganic nanoparticles are reported to have great gene delivery efficacy on surface modification. Chen et al. have discussed in great detail about the use of inorganic nanoparticle as a drug codelivery nanosystem [149]. Table 7 lists out some of the important nanoparticle based gene therapeutic approaches. Advancement in clinical studies of engineered siRNA-loaded nanoparticles has been discussed in Table 8 [150,151].

Table 7
Nanoparticle-mediated gene therapy for cancer treatment
Type of gene therapyDrugNanoparticles usedCancer type/cell lineEffectsRef.
Suicide gene therapy Plasmid DNA encoding saporin gene U11 peptide functionalized lipid-protamine-DNA nanoparticle Triple negative breast cancer (MDA-MB-231) Tumor size was found to be significantly reduced in in vivo mice model [152
Suicide gene therapy Plasmid DNA encoding Herpes simplex virus thymidine kinase (HSVtk) gene Poly(beta-amino ester) nanoparticles Pediatric brain tumors Increased median survival in in vivo mice model [153
siRNA-based therapy c-Myc siRNA Gold-PEG nanoparticles Adenocarcinoma Reduction in tumor size by 80% in vivo [154
siRNA-based therapy Akt1 siRNA Polyethylenimine based nanoparticle Mouse colon cancer Reduced tumor growth [155
siRNA-based therapy Polo-like kinase-1 siRNA (siPLK1) Hyaluronan containing lipid-based (cholesterol, DSPC, Dlin-MC3-DMA, DMG-PEG and DSPE-PEG amine) nanoparticle Glioblastoma Increased gene silencing efficiency and higher survival in mice [156
siRNA based therapy Cell division cycle-associated protein 1 (CDCA1) siRNA Lipid (PEG lipids, PEG-C-DMA lipids, D-Lin-DMA lipids, DSPC and PEG) nanoparticles Hepatocellular carcinoma Significant anticancer efficacy [157
siRNA-based therapy Cyclin targeting siRNA Peptide
MPG (ac-GALFLGFLGAAGS
TMGAWSQPKKKRKV-Cya)
(Cya : Cysteamide) 
HS68 fibroblasts, HeLa, PC3, MCF-7 and SCK3-Her2 Block cancer cell proliferation by efficient down-regulation of cyclin B1 levels [105
siRNA-based therapy ERK1/2 silencing siRNA Peptide
c[-Arg-DHis-Arg- DHis-Arg-Lys (Lys(linoleyl)2)-Arg- DHis-Arg-D His-Arg-Glu-]-Lys(FAM)-NH2 
Breast cancer (MDA-MB-231) Down-regulation of Erk1/2 gene level in TNBC cell line MDA-MB-231 [86
siRNA-based therapy Raf-1 siRNA Peptide based
Modified block copolymers-poly(ethylene glycol)- poly(ε-caprolactone)-Tat (GRKKRRQRRRG) (MPEG-PCL-Tat) 
C6 cells Cell death in rat glioma cells [106,158
siRNA-based therapy VEGF siRNA Peptide
KALA (WEAKLAKAL
AKALAKHLAKALAK
ALKACEA) 
PC-3 cells EGF sequence-specific gene inhibition in prostate carcinoma in vitro [107
siRNA-based therapy Polo like kinase-1 siRNA Multi-walled carbon nanotubes with amino functionalization Human lung carcinoma Significant regression of tumor volume [159
siRNA-based therapy Cyclin B1 siRNA and survivin siRNA Calcium phosphate nanoparticles Non-small cell lung cancer Significant gene silencing, reduction in cell growth and induction of apoptosis [160
miRNA-based therapy Tumor suppressor miR-31 and oncogenic miR-1323 Cysteamine functionalized gold nanoparticle Neuroblastoma and ovarian cancer Increased payload, efficient cellular uptake and reduced toxicity [161
miRNA-based therapy AntimiR-21 and antimiR-10b uPA peptide conjugated PLGA-b-PEG nanoparticles Triple negative breast cancer Reduction in tumor growth by 40% [162
shRNA-based therapy Doxorubicin encapsulated PLGA nanoparticle and Bcl-xL shRNA Alkyl modified polyethylenimine Breast cancer cell line MCF-7 Increased apoptosis of tumor cells and enhanced synergistic effect in comparison with only doxorubicin encapsulated PLGA nanoparticle treatment [163
shRNA-based therapy Akt1 shRNA Folate and chitosan grafted polyethylenimine copolymer Human lung carcinoma Enhanced cell transfection and reduced tumorigenesis [164
DNA vaccine Plasmid encoding prostate stem cell antigen Cationic RALA peptide/pDNA nanoparticles Prostate cancer Showed anticancer activity in vivo [165
mRNA vaccine mRNA encoding tumor associated antigen gp100 and TRP-2 Lipid based (DOTAP, DODAP, C12-200, cKK-E12, DOPE, DSPC, POPE, DMPC, DOPS, cholesterol, PEG, arachidonic acid, oleic acid, myristic acid) nanoparticle Melanoma (B16F10) Promoted enhanced cytotoxic T-cell response and reduced rate of tumor growth [166
Type of gene therapyDrugNanoparticles usedCancer type/cell lineEffectsRef.
Suicide gene therapy Plasmid DNA encoding saporin gene U11 peptide functionalized lipid-protamine-DNA nanoparticle Triple negative breast cancer (MDA-MB-231) Tumor size was found to be significantly reduced in in vivo mice model [152
Suicide gene therapy Plasmid DNA encoding Herpes simplex virus thymidine kinase (HSVtk) gene Poly(beta-amino ester) nanoparticles Pediatric brain tumors Increased median survival in in vivo mice model [153
siRNA-based therapy c-Myc siRNA Gold-PEG nanoparticles Adenocarcinoma Reduction in tumor size by 80% in vivo [154
siRNA-based therapy Akt1 siRNA Polyethylenimine based nanoparticle Mouse colon cancer Reduced tumor growth [155
siRNA-based therapy Polo-like kinase-1 siRNA (siPLK1) Hyaluronan containing lipid-based (cholesterol, DSPC, Dlin-MC3-DMA, DMG-PEG and DSPE-PEG amine) nanoparticle Glioblastoma Increased gene silencing efficiency and higher survival in mice [156
siRNA based therapy Cell division cycle-associated protein 1 (CDCA1) siRNA Lipid (PEG lipids, PEG-C-DMA lipids, D-Lin-DMA lipids, DSPC and PEG) nanoparticles Hepatocellular carcinoma Significant anticancer efficacy [157
siRNA-based therapy Cyclin targeting siRNA Peptide
MPG (ac-GALFLGFLGAAGS
TMGAWSQPKKKRKV-Cya)
(Cya : Cysteamide) 
HS68 fibroblasts, HeLa, PC3, MCF-7 and SCK3-Her2 Block cancer cell proliferation by efficient down-regulation of cyclin B1 levels [105
siRNA-based therapy ERK1/2 silencing siRNA Peptide
c[-Arg-DHis-Arg- DHis-Arg-Lys (Lys(linoleyl)2)-Arg- DHis-Arg-D His-Arg-Glu-]-Lys(FAM)-NH2 
Breast cancer (MDA-MB-231) Down-regulation of Erk1/2 gene level in TNBC cell line MDA-MB-231 [86
siRNA-based therapy Raf-1 siRNA Peptide based
Modified block copolymers-poly(ethylene glycol)- poly(ε-caprolactone)-Tat (GRKKRRQRRRG) (MPEG-PCL-Tat) 
C6 cells Cell death in rat glioma cells [106,158
siRNA-based therapy VEGF siRNA Peptide
KALA (WEAKLAKAL
AKALAKHLAKALAK
ALKACEA) 
PC-3 cells EGF sequence-specific gene inhibition in prostate carcinoma in vitro [107
siRNA-based therapy Polo like kinase-1 siRNA Multi-walled carbon nanotubes with amino functionalization Human lung carcinoma Significant regression of tumor volume [159
siRNA-based therapy Cyclin B1 siRNA and survivin siRNA Calcium phosphate nanoparticles Non-small cell lung cancer Significant gene silencing, reduction in cell growth and induction of apoptosis [160
miRNA-based therapy Tumor suppressor miR-31 and oncogenic miR-1323 Cysteamine functionalized gold nanoparticle Neuroblastoma and ovarian cancer Increased payload, efficient cellular uptake and reduced toxicity [161
miRNA-based therapy AntimiR-21 and antimiR-10b uPA peptide conjugated PLGA-b-PEG nanoparticles Triple negative breast cancer Reduction in tumor growth by 40% [162
shRNA-based therapy Doxorubicin encapsulated PLGA nanoparticle and Bcl-xL shRNA Alkyl modified polyethylenimine Breast cancer cell line MCF-7 Increased apoptosis of tumor cells and enhanced synergistic effect in comparison with only doxorubicin encapsulated PLGA nanoparticle treatment [163
shRNA-based therapy Akt1 shRNA Folate and chitosan grafted polyethylenimine copolymer Human lung carcinoma Enhanced cell transfection and reduced tumorigenesis [164
DNA vaccine Plasmid encoding prostate stem cell antigen Cationic RALA peptide/pDNA nanoparticles Prostate cancer Showed anticancer activity in vivo [165
mRNA vaccine mRNA encoding tumor associated antigen gp100 and TRP-2 Lipid based (DOTAP, DODAP, C12-200, cKK-E12, DOPE, DSPC, POPE, DMPC, DOPS, cholesterol, PEG, arachidonic acid, oleic acid, myristic acid) nanoparticle Melanoma (B16F10) Promoted enhanced cytotoxic T-cell response and reduced rate of tumor growth [166
Table 8
Nanocarrier-mediated siRNA delivery systems in clinical trials
DrugDiseaseTargetNanoparticleCompanyStatus
Phase I 
CALAA-01 Solid tumors RRM 2 (RNA recognition motif domain of the Rbfox family protein) Cyclodextrin/PEG, transferrin Calando Pharm Terminated 
ALN-VSP02 Solid tumors with liver lesion VEGF, KSP (kinesin spindle protein) SNALP (stable nucleic acid-lipid particles) Alnylam Pharm. Completed 
siRNA–EphA2–DOPC Advanced cancer EphA2 (tyrosine kinase)(a key effector of the MEK/ERK/RSK pathway) Liposome M.D. Anderson Cancer Center Not completed yet 
Phase II 
FANG Ovarian tumors FURIN (a protease enzyme) Liposome Gradalis, Inc. Active 
Atu027 Advanced or metastatic solid tumors PKN3 (protein kinase N3) Liposome Silence Therapeutics Completed 
siG12D LODER Pancreatic ductal adenocarcinoma KRASG12D Polymer matrix Silenseed Ltd. Ongoing 
TKM- 080301 or TKM-PLK1 Solid tumors PLK1 SNALP Tekmira Pharma Completed 
DCR-MYC Hepatocellular carcinoma, solid tumors, non-Hodgkins lymphoma, multiple myeloma, pancreatic neuroendocrine tumors MYC Systemic/IV infusion Dicerna Pharmaceuticals Terminated 
DrugDiseaseTargetNanoparticleCompanyStatus
Phase I 
CALAA-01 Solid tumors RRM 2 (RNA recognition motif domain of the Rbfox family protein) Cyclodextrin/PEG, transferrin Calando Pharm Terminated 
ALN-VSP02 Solid tumors with liver lesion VEGF, KSP (kinesin spindle protein) SNALP (stable nucleic acid-lipid particles) Alnylam Pharm. Completed 
siRNA–EphA2–DOPC Advanced cancer EphA2 (tyrosine kinase)(a key effector of the MEK/ERK/RSK pathway) Liposome M.D. Anderson Cancer Center Not completed yet 
Phase II 
FANG Ovarian tumors FURIN (a protease enzyme) Liposome Gradalis, Inc. Active 
Atu027 Advanced or metastatic solid tumors PKN3 (protein kinase N3) Liposome Silence Therapeutics Completed 
siG12D LODER Pancreatic ductal adenocarcinoma KRASG12D Polymer matrix Silenseed Ltd. Ongoing 
TKM- 080301 or TKM-PLK1 Solid tumors PLK1 SNALP Tekmira Pharma Completed 
DCR-MYC Hepatocellular carcinoma, solid tumors, non-Hodgkins lymphoma, multiple myeloma, pancreatic neuroendocrine tumors MYC Systemic/IV infusion Dicerna Pharmaceuticals Terminated 

Combination therapy, a treatment modality involving combination of two or more therapeutic agents, has now become the cornerstone of cancer therapy since single drug based monotherapy failed to provide a considerable therapeutic response. Enhanced therapeutic potential, reduced toxicity and prevention of drug resistance mediated by combination therapy have hugely increased the therapeutic response of cancer patients. Combination therapy can be designed in two different ways. First, targeting of different molecular pathways by multiple drugs can lead to delayed cellular adaptation and oncogenic mutations. Second, targeting the same pathway could be helpful in the development of synergistic interaction of multiple drugs with higher efficacy and target selectivity. Synergistic interaction is best described by the Chou-Talalay method where combination index (CI) is less than 1 [167,168]. Boshuizen et al. suggested that a synergistic drug interaction in a combination therapy can be rationally designed by invoking several principles: (1) multiple targeting of a signaling pathway, (2) maximal driver pathway inhibition, (3) targeting agents responsible for signal reactivation, (4) enhanced synthetic lethality, (5) targeting heterogeneous and drug resistant cell populations in a tumor, (6) targeting immune cell function and tumor microenvironment modulation and (7) neoadjuvant therapies [169]. Combination therapy also employs repurposing of drugs that were initially used for diseases other than cancer. This approach saves the high cost and time required for granting FDA approval as the repurposed drug is generally an FDA approved drug, thus cutting the cost of the treatment [170]. The synergistic agents involved in modern combination therapies should ideally have a different pharmacological mechanism of action, exhibit no cross-resistance or overlapping toxicities and target tumor heterogeneity [171]. By integrating the knowledge and progress made in the field of mechanisms responsible for tumorigenesis, tumor microenvironment, therapy response and cancer heterogeneity, an effective treatment can be designed [169].

Nanoparticles have emerged itself as a potent tool to be used in combination therapy as they provide longer circulation time of biologically active drug, reduced toxicity, improved drug solubility, controlled release and has specific target potential [172]. Gurunathan and co-workers have elaborately described different types of nanoparticles mediated combination therapies [173]. The rationale for designing nanoparticle-mediated combination therapy for cancer treatment is provided in Figure 9. In Table 9, we have enlisted examples of nanoparticle-mediated combination therapy for treating different types of cancer and their advantages over monotherapy.

Rationale of nanoparticle-mediated combination therapy for cancer

Figure 9
Rationale of nanoparticle-mediated combination therapy for cancer

Combination therapy has been divided according to the target of action and mode of delivery. The figure represents the individual advantages of targeting same and different signaling pathways and the advantages of co-delivery over free drug delivery.

Figure 9
Rationale of nanoparticle-mediated combination therapy for cancer

Combination therapy has been divided according to the target of action and mode of delivery. The figure represents the individual advantages of targeting same and different signaling pathways and the advantages of co-delivery over free drug delivery.

Close modal
Table 9
Nanoparticle-mediated combination therapy with small molecules and nucleotide-based anticancer drugs
Drug 1Mode of action of drug 1Drug 2Mode of action of drug 2IndicationComments on combinationRef.
Sterically stabilized liposomal DOX Cytotoxic Liposome containing Bcl-2 antisense oligodeoxynucleotide, G3139 Gene silencing In vivo melanoma model Combination showed delayed tumor growth and increased accumulation of DOX in tumor site than monotherapy [174
Liposomal daunorubicin Antitumor antibiotic Cytarabine Antineoplastic anti-metabolite Patients having refractory or recurring acute myeloid leukemia The combination has significant antileukemia activity with low toxicity. Liposomal encapsulation of daunorubicin changes the pharmacology profile to decrease toxicity and increase delivery to tumor sites [175
Liposome-entrapped, ends-modified raf antisense oligonucleotide (LErafAON) Gene silencing Cisplatin/epirubicin/mitoxantrone/docetaxel/gemcitabine Chemotherapeutic agents In vivo pancreas or pancreatic cancer model Increased tumor growth inhibition as compared with single agents [176
aGD2-SIL(DOX) Sterically stabilized immunoliposomes(SIL) encapsulated with DOX, targeted to the disialoganglioside receptor GD2 Cytotoxic NGR-SL(DOX) Sterically stabilized immunoliposomes (SIL) encapsulated with DOX, targeted to angiogenic endothelial cell marker aminopeptidase N by peptide NGR Cytotoxic In vivo neuroblastoma model Considerable reduction of the angiogenic potential of various neuroblastoma xenografts [177
Non-pegylated liposomal Doxorubicin Cytotoxic Cyclophosphamide/ docetaxel Cytotoxic Patients with metastatic breast cancer Use of non-pegylated liposomal Doxorubicin in combination with other drugs can be used for the first-line therapy against metastatic breast cancer [178
RGD SSL-DOX (RGD-SSL- RGD-modified sterically stabilized liposomes) Cytotoxic RGD-lipo-siRNA silencing P-glycoprotein Gene silencing In vivo mouse model of drug-resistant MCF7/A breast cancer tumor Combination exhibited higher in vitro toxicity and greater inhibition of tumor growth [179
VEGF siRNA in polycation liposome-encapsulated calcium phosphate nanoparticles Gene silencing Doxorubicin Cytotoxic In vivo mouse model of breast cancer Combination showed significant tumor growth and angiogenesis inhibition [180
Polymeric nanoparticle- encapsulated curcumin Cytotoxic Gemcitabine Cytotoxic In vivo mouse model of human pancreatic cancer Enhanced tumor growth inhibition compared to single agents. [181
VOR-POEOMA Vorinostat encapsulated into poly(ethylene glycol) monomethacrylate
(POEOMA)-based disulfide cross-linked nanogels. 
Histone deacetylase inhibitor ETOP-POEOMA
Etopside encapsulated into poly(ethylene glycol) monomethacrylate
(POEOMA)-based disulfide cross-linked nanogels. 
Topoisomerase II inhibitor Human cervical HeLa cancer cells Combination showed enhanced synergistic cell killing efficiency [182
C60 fullerene Cytotoxic Doxorubicin Cytotoxic In vivo mouse model of lung cancer Combination resulted in increased apoptosis in tumor cells and tumor growth inhibition [183
Anti Bcl-2 siRNA loaded polyethylenimine (PEI)-conjugated graphene oxide (PEI-GO) Gene silencing DOX loaded polyethylenimine (PEI)-conjugated graphene oxide (PEI-GO) Cytotoxic Human cervical HeLa cancer cells Sequential delivery exhibited synergistic effect. Codelivery showed no significant synergistic effect on killing cancer cells [184
Palladium nanoparticles (PdNPs) Cytotoxic Trichostatin A (TSA) Histone deacetylase inhibitor Human cervical HeLa cancer cells Combination exhibited synergistic interaction and also had an increased effect on cytotoxicity, oxidative stress and caspase-3/9 activity [185
Palladium nanoparticles (PdNPs) Cytotoxic Tubastatin A (TUB-A) Histone deacetylase inhibitor TNBC cell line MDA-MB-231 Combination had a more pronounced effect on the inhibition of HDAC activity and enhanced apoptosis of cells [186
Drug 1Mode of action of drug 1Drug 2Mode of action of drug 2IndicationComments on combinationRef.
Sterically stabilized liposomal DOX Cytotoxic Liposome containing Bcl-2 antisense oligodeoxynucleotide, G3139 Gene silencing In vivo melanoma model Combination showed delayed tumor growth and increased accumulation of DOX in tumor site than monotherapy [174
Liposomal daunorubicin Antitumor antibiotic Cytarabine Antineoplastic anti-metabolite Patients having refractory or recurring acute myeloid leukemia The combination has significant antileukemia activity with low toxicity. Liposomal encapsulation of daunorubicin changes the pharmacology profile to decrease toxicity and increase delivery to tumor sites [175
Liposome-entrapped, ends-modified raf antisense oligonucleotide (LErafAON) Gene silencing Cisplatin/epirubicin/mitoxantrone/docetaxel/gemcitabine Chemotherapeutic agents In vivo pancreas or pancreatic cancer model Increased tumor growth inhibition as compared with single agents [176
aGD2-SIL(DOX) Sterically stabilized immunoliposomes(SIL) encapsulated with DOX, targeted to the disialoganglioside receptor GD2 Cytotoxic NGR-SL(DOX) Sterically stabilized immunoliposomes (SIL) encapsulated with DOX, targeted to angiogenic endothelial cell marker aminopeptidase N by peptide NGR Cytotoxic In vivo neuroblastoma model Considerable reduction of the angiogenic potential of various neuroblastoma xenografts [177
Non-pegylated liposomal Doxorubicin Cytotoxic Cyclophosphamide/ docetaxel Cytotoxic Patients with metastatic breast cancer Use of non-pegylated liposomal Doxorubicin in combination with other drugs can be used for the first-line therapy against metastatic breast cancer [178
RGD SSL-DOX (RGD-SSL- RGD-modified sterically stabilized liposomes) Cytotoxic RGD-lipo-siRNA silencing P-glycoprotein Gene silencing In vivo mouse model of drug-resistant MCF7/A breast cancer tumor Combination exhibited higher in vitro toxicity and greater inhibition of tumor growth [179
VEGF siRNA in polycation liposome-encapsulated calcium phosphate nanoparticles Gene silencing Doxorubicin Cytotoxic In vivo mouse model of breast cancer Combination showed significant tumor growth and angiogenesis inhibition [180
Polymeric nanoparticle- encapsulated curcumin Cytotoxic Gemcitabine Cytotoxic In vivo mouse model of human pancreatic cancer Enhanced tumor growth inhibition compared to single agents. [181
VOR-POEOMA Vorinostat encapsulated into poly(ethylene glycol) monomethacrylate
(POEOMA)-based disulfide cross-linked nanogels. 
Histone deacetylase inhibitor ETOP-POEOMA
Etopside encapsulated into poly(ethylene glycol) monomethacrylate
(POEOMA)-based disulfide cross-linked nanogels. 
Topoisomerase II inhibitor Human cervical HeLa cancer cells Combination showed enhanced synergistic cell killing efficiency [182
C60 fullerene Cytotoxic Doxorubicin Cytotoxic In vivo mouse model of lung cancer Combination resulted in increased apoptosis in tumor cells and tumor growth inhibition [183
Anti Bcl-2 siRNA loaded polyethylenimine (PEI)-conjugated graphene oxide (PEI-GO) Gene silencing DOX loaded polyethylenimine (PEI)-conjugated graphene oxide (PEI-GO) Cytotoxic Human cervical HeLa cancer cells Sequential delivery exhibited synergistic effect. Codelivery showed no significant synergistic effect on killing cancer cells [184
Palladium nanoparticles (PdNPs) Cytotoxic Trichostatin A (TSA) Histone deacetylase inhibitor Human cervical HeLa cancer cells Combination exhibited synergistic interaction and also had an increased effect on cytotoxicity, oxidative stress and caspase-3/9 activity [185
Palladium nanoparticles (PdNPs) Cytotoxic Tubastatin A (TUB-A) Histone deacetylase inhibitor TNBC cell line MDA-MB-231 Combination had a more pronounced effect on the inhibition of HDAC activity and enhanced apoptosis of cells [186

In recent years, there has been an increase in the number of reports of multifunctional nanoparticle mediated combination therapy aided with peptides. Some of the advantages of peptide-mediated combination nanotherapeutics are tumor targeted delivery, presentation of tumor antigens for elicited immune response, sensitization of drug resistant cells and reduced side effects [187]. Mehrotra et al. and Mallick et al. have elaborately enlisted different types of peptide-based combination nanotherapeutics [187,188]. We have enlisted some of them in Table 10.

Table 10
Nanoparticle-mediated combination therapy having peptide-based anticancer drug
Peptide + Combination drugDrug detailsIndicationsCommentsRef.
dPPA peptide + paclitaxel prodrug + pheophorbide A dPPA-1 peptide –NYSKPTDRQYHF (anti-PD-L1) paclitaxel- cytotoxic pheophorbide A- photosensitier In vivo, breast cancer and lung metastasis Increased NK cell and T-cell activation causing inhibition of complete lung metastasis and at least 10% decrease in primary tumor volume as compared with either alone or combination of paclitaxel and pheophorbideA [189
NuBCP-9 (Bcl-2 inhibitor) peptide + paclitaxel NuBCP-9 -Ac-FSRSLHSLLGC-NH2 paclitaxel-cytotoxic In vivo, breast cancer Combination leads to reduced IC50 value (100-fold) in paclitaxel resistant cells and shows complete tumor inhibition in syngeneic mice model as compared with only paclitaxel [190,191
Acetylated rapeseed protein isolate derived peptides + DOX From hydrolyzed ARPI peptides 3 bioactive peptides were screened. Sequences are AGS, PAS and YT. DOX- cytotoxic In vivo, breast cancer Enhanced cellular uptake and nuclear transport in comparison with free DOX. Increase in tumor inhibition and diminished DOX-associated cardiotoxicity. [192
PMI + BIM (Bcl-2 inhibitor) peptide + iNGR PMI- p53 activating) peptide- TSFAEYWNLLSP BIM- Bcl-2 inhibitor) peptide-MRPEIWIAQELRRIGDEFNAYYARRV iNGR- CD-13) targeting peptide cyclic CRNGRGPDC In vivo, colorectal cancer Increased tumor inhibition (15%) with significant improvement in biosafety and reduced body weight loss compared with only DOX. [193,194
aFLT1 peptide + DOX aFLT1 peptide- binds VEGFR1 isoform- GNQWFI-NH2 DOX-cytotoxic In vivo, breast cancer Two-fold increase in tumor inhibition [195
KLA + DOX KLA - mitochondrial membrane disruptor) peptide KLAKLAKKLAKLAK DOX-cytotoxic In vivo, colorectal cancer Marked increase in tumor inhibition and mean survival time as compared with only DOX [196
R8 modified AVPI peptide with p53DNA+DOX R8 modified AVPI peptide- cell penetrating apoptotic peptide- AVPIR8 p53 DNA- induces apoptosis DOX-cytotoxic In vivo, resistant breast cancer model 4.4- and 2-fold increase in tumor inhibition in drug resistant mouse model as compared with equal and high free DOX dose, respectively [197
KLA peptide + chlorin e6 KLA peptide- membrane lysis peptide-D-(KLAKLAK)2 chlorin e6- generates singlet oxygen causing membrane disruption In vitro, cervical carcinoma Tenfold reduced IC50 value compared to only peptide [198
Wilms tumor gene (WT1) peptide-based vaccine+gemcitabine WT1 peptide- target antigens for cancer immunotherapy CYTWNQMNL Gemcitabine- cytotoxic Patients with advanced pancreatic cancer Combination was found to be more effective than gemcitabine alone and combination therapy was well tolerated [188,199
Cep55/c10orf3_193+ Cep55/c10orf3_402 + Cep55/c10orf3_283 VYVKGLLAKI + EFAITEPLVTF + LYSQRRADVQHL antigenic peptides Patients with colorectal carcinoma Vaccination involving peptide mixture could be more efficacious compared with single peptide to treat colorectal carcinoma patients [188,200
“Peptide cocktail” RNF43–721: NSQPVWLCL
TOMM34–299: KLRQEVKQNL
KOC1(IMP-3)-508 (KTVNELQNL)
3 peptides derived from oncoantigens
VEGFR1–1084: SYGVLLWEI
VEGFR2–169: RFVPDGNRI
2 peptides derived from angiogenesis factors 
Patients with advanced colorectal cancer Treatment with multiple peptides was well tolerated without systemic adverse effects. The median overall survival time was 13.5 months [188,201
7-peptide cocktail vaccine RNF43: NSQPVWLCL
TOMM34: KLRQEVKQNL
FOXM1: IYTWIEDHF
MELK: EYCPGGNLF
HJURP: KWLISPVKI
5 tumor antigen-derived peptides
VEGFR1: SYGVLLWEI
VEGFR2: RFVPDGNRI
2 vascular endothelial growth factor receptor-derived peptides 
Patients with metastatic colorectal cancer Patients exhibiting positive cytotoxic T lymphocyte responses to all seven peptides had longer overall survival compared with other patients and this therapy is recommended for further trials [188,202
KIF20A-derived peptide + gemcitabine KIF20A-peptide for trafficking of molecules and organelles during the growth of pancreatic cancer- KVYLRVRPLL
Gemcitabine- cytotoxic 
Patients with advanced pancreatic cancer The disease control rate was 44%. The median survival time after first vaccination was 173 days and 1-year survival rate was 11.1%. No severe adverse effects of grade 3 or higher were observed [188,203
GV1001 + gemcitabine GV1001 - telomerase peptide- EARPALLTSRLRFIPK
Gemcitabine- cytotoxic 
Patients with advanced pancreatic cancer This combination appears to be safer with transient and weak immune responses [188,204
E75 + GM-CSF E75- immunogenic peptide derived from the HER2 protein. KIFGSLAFL
GM-CSF = granulocyte-macrophage colony-stimulating factor 
Patients with node-positive or high-risk node-negative breast cancer Therapy considered safe with a suggestion of clinical benefit. Has been licensed for commercial development [188,205
FNIII14+ Ara C FNIII14-peptide derived from fibronectin- TEATITGLEPGTEYTIYVIAL
Ara C – anti metabolic agent cytarabine/arabinosylcytosine 
In vivo minimum residual disease (MRD) mice model In mouse with MRD in bone marrow, 100% survival was achieved with this combination, whereas Ara C alone prolonged survival only slightly [188,206,207
D-K6L 9 + IL-12 D-K6L9 - induces necrosis in cancer cells-Ac[D(K6L9)]-NH2
IL-12- pro-inflammatory cytokine-interleukin 12 
In vivo murine melanoma model This combination showed long-term tumor growth inhibitory effect [188,208
VEGFR2–169 + S-1 + cisplatin VEGFR2–169-RFVPDGNRI
S-1 - combination drug tegafur/gimeracil/oteracil cisplatin - cytotoxic 
Patients with advanced gastric cancer The combination therapy was highly effective and well tolerated in advanced or recurrent gastric cancer [188,209
LD8 + DOX LD8 – gramicidin A inspired peptide Boc-LA-DV-LL-DA-LV-DA-LL-DW- OMe
DOX- cytotoxic 
In vitro TNBC cell line MDA-MB-231 LD8-DOX-NP induces G2 phase cell cycle arrest and apoptosis of MDA-MB-231 [210
Peptide + Combination drugDrug detailsIndicationsCommentsRef.
dPPA peptide + paclitaxel prodrug + pheophorbide A dPPA-1 peptide –NYSKPTDRQYHF (anti-PD-L1) paclitaxel- cytotoxic pheophorbide A- photosensitier In vivo, breast cancer and lung metastasis Increased NK cell and T-cell activation causing inhibition of complete lung metastasis and at least 10% decrease in primary tumor volume as compared with either alone or combination of paclitaxel and pheophorbideA [189
NuBCP-9 (Bcl-2 inhibitor) peptide + paclitaxel NuBCP-9 -Ac-FSRSLHSLLGC-NH2 paclitaxel-cytotoxic In vivo, breast cancer Combination leads to reduced IC50 value (100-fold) in paclitaxel resistant cells and shows complete tumor inhibition in syngeneic mice model as compared with only paclitaxel [190,191
Acetylated rapeseed protein isolate derived peptides + DOX From hydrolyzed ARPI peptides 3 bioactive peptides were screened. Sequences are AGS, PAS and YT. DOX- cytotoxic In vivo, breast cancer Enhanced cellular uptake and nuclear transport in comparison with free DOX. Increase in tumor inhibition and diminished DOX-associated cardiotoxicity. [192
PMI + BIM (Bcl-2 inhibitor) peptide + iNGR PMI- p53 activating) peptide- TSFAEYWNLLSP BIM- Bcl-2 inhibitor) peptide-MRPEIWIAQELRRIGDEFNAYYARRV iNGR- CD-13) targeting peptide cyclic CRNGRGPDC In vivo, colorectal cancer Increased tumor inhibition (15%) with significant improvement in biosafety and reduced body weight loss compared with only DOX. [193,194
aFLT1 peptide + DOX aFLT1 peptide- binds VEGFR1 isoform- GNQWFI-NH2 DOX-cytotoxic In vivo, breast cancer Two-fold increase in tumor inhibition [195
KLA + DOX KLA - mitochondrial membrane disruptor) peptide KLAKLAKKLAKLAK DOX-cytotoxic In vivo, colorectal cancer Marked increase in tumor inhibition and mean survival time as compared with only DOX [196
R8 modified AVPI peptide with p53DNA+DOX R8 modified AVPI peptide- cell penetrating apoptotic peptide- AVPIR8 p53 DNA- induces apoptosis DOX-cytotoxic In vivo, resistant breast cancer model 4.4- and 2-fold increase in tumor inhibition in drug resistant mouse model as compared with equal and high free DOX dose, respectively [197
KLA peptide + chlorin e6 KLA peptide- membrane lysis peptide-D-(KLAKLAK)2 chlorin e6- generates singlet oxygen causing membrane disruption In vitro, cervical carcinoma Tenfold reduced IC50 value compared to only peptide [198
Wilms tumor gene (WT1) peptide-based vaccine+gemcitabine WT1 peptide- target antigens for cancer immunotherapy CYTWNQMNL Gemcitabine- cytotoxic Patients with advanced pancreatic cancer Combination was found to be more effective than gemcitabine alone and combination therapy was well tolerated [188,199
Cep55/c10orf3_193+ Cep55/c10orf3_402 + Cep55/c10orf3_283 VYVKGLLAKI + EFAITEPLVTF + LYSQRRADVQHL antigenic peptides Patients with colorectal carcinoma Vaccination involving peptide mixture could be more efficacious compared with single peptide to treat colorectal carcinoma patients [188,200
“Peptide cocktail” RNF43–721: NSQPVWLCL
TOMM34–299: KLRQEVKQNL
KOC1(IMP-3)-508 (KTVNELQNL)
3 peptides derived from oncoantigens
VEGFR1–1084: SYGVLLWEI
VEGFR2–169: RFVPDGNRI
2 peptides derived from angiogenesis factors 
Patients with advanced colorectal cancer Treatment with multiple peptides was well tolerated without systemic adverse effects. The median overall survival time was 13.5 months [188,201
7-peptide cocktail vaccine RNF43: NSQPVWLCL
TOMM34: KLRQEVKQNL
FOXM1: IYTWIEDHF
MELK: EYCPGGNLF
HJURP: KWLISPVKI
5 tumor antigen-derived peptides
VEGFR1: SYGVLLWEI
VEGFR2: RFVPDGNRI
2 vascular endothelial growth factor receptor-derived peptides 
Patients with metastatic colorectal cancer Patients exhibiting positive cytotoxic T lymphocyte responses to all seven peptides had longer overall survival compared with other patients and this therapy is recommended for further trials [188,202
KIF20A-derived peptide + gemcitabine KIF20A-peptide for trafficking of molecules and organelles during the growth of pancreatic cancer- KVYLRVRPLL
Gemcitabine- cytotoxic 
Patients with advanced pancreatic cancer The disease control rate was 44%. The median survival time after first vaccination was 173 days and 1-year survival rate was 11.1%. No severe adverse effects of grade 3 or higher were observed [188,203
GV1001 + gemcitabine GV1001 - telomerase peptide- EARPALLTSRLRFIPK
Gemcitabine- cytotoxic 
Patients with advanced pancreatic cancer This combination appears to be safer with transient and weak immune responses [188,204
E75 + GM-CSF E75- immunogenic peptide derived from the HER2 protein. KIFGSLAFL
GM-CSF = granulocyte-macrophage colony-stimulating factor 
Patients with node-positive or high-risk node-negative breast cancer Therapy considered safe with a suggestion of clinical benefit. Has been licensed for commercial development [188,205
FNIII14+ Ara C FNIII14-peptide derived from fibronectin- TEATITGLEPGTEYTIYVIAL
Ara C – anti metabolic agent cytarabine/arabinosylcytosine 
In vivo minimum residual disease (MRD) mice model In mouse with MRD in bone marrow, 100% survival was achieved with this combination, whereas Ara C alone prolonged survival only slightly [188,206,207
D-K6L 9 + IL-12 D-K6L9 - induces necrosis in cancer cells-Ac[D(K6L9)]-NH2
IL-12- pro-inflammatory cytokine-interleukin 12 
In vivo murine melanoma model This combination showed long-term tumor growth inhibitory effect [188,208
VEGFR2–169 + S-1 + cisplatin VEGFR2–169-RFVPDGNRI
S-1 - combination drug tegafur/gimeracil/oteracil cisplatin - cytotoxic 
Patients with advanced gastric cancer The combination therapy was highly effective and well tolerated in advanced or recurrent gastric cancer [188,209
LD8 + DOX LD8 – gramicidin A inspired peptide Boc-LA-DV-LL-DA-LV-DA-LL-DW- OMe
DOX- cytotoxic 
In vitro TNBC cell line MDA-MB-231 LD8-DOX-NP induces G2 phase cell cycle arrest and apoptosis of MDA-MB-231 [210

However, the current combination therapy treatment in cancer, especially in the case of metastatic breast cancer, is still loaded with flaws having moderate efficacy but additive toxicity [18]. Drugs, when administered separately without any modification, tend to not only cause an additive anticancer effect but also result in augmented adverse effects of each drug as well. For achieving synergistic interactions, a definite ratio of the two free drugs needs to be maintained. This ratio is generally determined in in vitro studies, and it is crucial that this ratio is to be maintained at tumor site. The problem arises because in in vivo model it becomes virtually impossible to deliver the determined ratio of free drugs in the tumor site. This is mainly due to the different pharmacokinetic properties, elimination and metabolism rates of individual drugs. Delivery of non-fixed ratio of drugs can give rise to antagonistic interactions and drug resistance in cancer cells [257]. For example, irinotecan and cisplatin (both cytotoxic drugs) show synergistic interaction at 4:1 ratio but strong antagonistic interaction at 1:1 ratio and in the case of many anticancer drugs, when the effective dose is below its optimum dose, it may give rise to drug resistant cells in tumor [257]. Another complication arises when the free drugs need to be administered by different routes and at different schedules. HER2 targeted combination therapy with Trastuzumab (TRZ, a monoclonal antibody binding to HER2 receptor) and lapatinib (a tyrosine kinase inhibitor blocking HER2 and EGFR pathways) have two different routes of administration. Lapatinib is administered daily as an oral formulation, while TRZ is given weekly as an intravenous drug. This difference in schedules and ways of administration of these two drugs makes the management of pharmacokinetic and pharmacodynamic profiles more challenging and virtually impossible to achieve uniform temporal and spatial co-delivery [18]. Most standard chemotherapy guidelines prefer sequential delivery of free drugs over concurrent delivery due to toxicity issues. This also prevents the right temporal delivery of drugs with dissimilar pharmacokinetic properties [258]. To address these issues, nanoparticle-mediated co-delivery of those drugs may ensure the desired spatio-temporal delivery and controlled release of drugs while maintaining the synergistic fixed ratio (Figure 10) [258]. Nanocarrier mediated co-encapsulated drugs having physically different properties show similar pharmacokinetic profiles, extended drug half-life, solubility, tumor accumulation and synergistic drug interactions when compared with free drugs administered sequentially or concurrently [18,257]. Cytrabine (an antimetabolite chemotherapeutic drug) and danorubicin (a cytotoxic chemotherapeutic drug) has distinct pharmacokinetic properties as free drugs but when co-encapsulated in liposomal nanocarrier as Vyxeos® exhibit similar pharmacokinetic profile [257]. Nanocarrier mediated co-delivery of drugs enables targeting multiple signaling pathways, overcoming drug resistance and immunosuppression by cancer [257].

Schematic representation of nanoparticle-mediated ratiometric delivery of drug combination

Figure 10
Schematic representation of nanoparticle-mediated ratiometric delivery of drug combination

The scheme represents the pharmacokinetics and biodistribution of ratiometric drug combination. Drug combination delivered sequentially or concurrently either by similar or different route of administration show a non-fixed ratio in biodistribution of both the drugs. Nanoparticle mediated delivery of dual drugs maintain a fixed ratio of biodistribution of drug combinations resulting in higher therapeutic efficacy (This image was drawn based on the information provided in Zhang et al. 2016 [258], Figure 4). Adapted from “Body (female, teen)”, by BioRender.com (2021). Retrieved from https://app.biorender.com/illustrations/61d7dc25883c8d00a22cf5c8.

Figure 10
Schematic representation of nanoparticle-mediated ratiometric delivery of drug combination

The scheme represents the pharmacokinetics and biodistribution of ratiometric drug combination. Drug combination delivered sequentially or concurrently either by similar or different route of administration show a non-fixed ratio in biodistribution of both the drugs. Nanoparticle mediated delivery of dual drugs maintain a fixed ratio of biodistribution of drug combinations resulting in higher therapeutic efficacy (This image was drawn based on the information provided in Zhang et al. 2016 [258], Figure 4). Adapted from “Body (female, teen)”, by BioRender.com (2021). Retrieved from https://app.biorender.com/illustrations/61d7dc25883c8d00a22cf5c8.

Close modal

Various delivery systems widely used for co-delivery of two drugs include liposomes, dendrimers, polymeric nanoparticles and water-soluble polymer conjugates. Lee et al. and Gurunathan et al. have elaborately reviewed nanoparticle-mediated co-delivery of two or more drugs for cancer therapy as described in Table 11 [18,173].

Table 11
Nanoparticle mediated co-delivery of drugs for cancer therapy
Carrier compositionTherapeuticsIndicationStatusTargetingRef.
Liposome based co-delivery of drugs against cancer 
PEG-Liposome Topotecan + Vincristine Brain cancer In vivo Passive [18,211
Polymer-caged nanobins (PCN); liposome surrounded by cholesterol-terminated poly(acrylic acid) Cisplatin + Doxorubicin Various cancers In vitro Passive [18
Liposome Cytarabine + Daunorubicin Acute myeloid leukemia Phase II Passive [18,212
Liposome Irinotecan + Floxuridine Colorectal cancer Phase II Passive [18,213,214
The mixture of two Liposomes Irinotecan + Cisplatin Small-cell lung cancer In vivo Passive [18,214
PEG-Liposome Quercetin + Vincristine Hormone- and TRZ insensitive breast cancer In vivo Passive [18,215
Cationic, anionic PEG Liposome VEGF/ c-myc siRNA + Doxorubicin MDR-breast cancer In vivo Passive [18,216
Liposome 6-Mercaptopurine + Daunorubicin Acute myeloid leukemia In vitro Passive [18,217
Dendrimer based co-delivery of drugs against cancer 
G5 PAMAM dendrimer (G5-Generation 5, PAMAM-poly(amidoamine)) Antisense-miRNA21 +5-fluorouracil Glioblastoma In vitro Active; miRNA overexpression [18,218
Aptamer-G4 PAMAM dendrimer conjugates (G4-Generation 4) Unmethylated CpG-oligonucleotides+ Doxorubicin Prostate cancer In vivo Active; a single-strand DNA-A9 prostate-specific membrane antigen, RNA aptamer hybrid [18,219
Dendritic PEG H2N–PEG–dendrimer– (COOH)4 Paclitaxel + alendronate Cancer bone metastasis In vivo Both passive and active Active by alendronate molecule [18,220
RGDfK-G3 Poly-lysine dendrimer (G3-Generation 3) Doxorubicin + siRNA Glioblastoma In vitro Active; αvβ3 integrin [18,221
Folate-G5 poly -propyleneimine dendrimer with ethylenediamine core (G5-Generation 5) Methotrexate + all-trans-retinoic acid Leukemia In vitro Active; folate receptor [18,222
Polymer based co-delivery of drugs against cancer 
PEG-PLGA Lonidamine + Paclitaxel Multiple drug resistant (MDR) breast cancer In vitro Active; EGFR [18,223
Methoxy PEG-PLGA Doxorubicin+ paclitaxel Various cancers In vitro Passive [18,223
PEG-PLA (PLA-poly(d,l, lactic acid)) Paclitaxel, Etoposide, or Docetaxel + 17-AAG Various cancers In vitro Active; HSP90 [18,224
PEG-PLA Combretastatin A4 + Doxorubicin Various cancers In vitro Active; angiogenesis [18,225
PDMAEMA-PCL- PDMAEMA poly(N,N-dimethylamino-2- ethyl methacrylate)- polycaprolactone-poly (N,N-dimethylamino- 2-ethyl methacrylate) Paclitaxel + VEGF siRNA Prostate cancer In vitro Active; VEGF [18,226
PEG-DSPE/PLGA Combretastatin + Doxorubicin Lung carcinoma In vitro Passive [18,115
PEG-PLA and PEG-DSPE/TPGS (TPGS- tocopheryl polyethylene glycol) Paclitaxel + 17-AAG(HSP90 inhibitor) Ovarian cancer In vitro Active; HSP90 [18,227
P(MDS-co-CES) poly (N-methyldietheneamine sebacate)- co-[(cholesteryl oxocarbonylamido ethyl) methyl bis(ethylene) ammonium bromide] Paclitaxel + Interleukin-12 or Bcl-2 siRNA Breast cancer In vivo Active; Bcl-2 [18,228
PEG-b-PHSA PEG-block-poly(N-hexyl stearate l-aspartamide) Doxorubicin + Wortmannin Breast cancer In vitro Passive [18,229
PLN formulation ((DG)n) (Polymer lipid hybrid nanoparticles (NP) co-loaded with DOX and GG918) Doxorubicin + GG918 Breast cancer In vitro Passive [230
PLGA Vincristine + Verapamil Hepatocellular carcinoma In vitro Passive [18,231
PLGA Paclitaxel + Tariquidar Breast cancer In vivo Active; functionalized with biotin [173,232
PLGA Rapamycin + piperine Breast cancer In vitro Passive [173,233
PACA polyalkylcyanoacrylate Doxorubicin + Cyclosporine A Various cancers In vitro Passive [18,234
PEG outer shell, middle PCL and inner CPCL core (PCL- polycaprolactone; CPCL- carboxylic functionalized PCL) Doxorubicin +Cisplatin Breast cancer In vitro Passive [173,235
Bradykinin-potentiating peptide decorated chitosan nanoparticle Bradykinin-potentiating peptide + bioreductively sensitive platinum (IV) compound which becomes cisplatin in intracellular reductive environment Hepatocellular carcinoma In vivo Passive [188,236
Polymeric micelles based co-delivery of drugs against cancer 
MPEG-b-P(LA-co-MCC) (MPEG-b-P(LA-co-MCC)) - methoxy poly(ethylene glycol)-block-poly(1-lactide -co-2-methyl-2- carboxyl-propylene carbonate) Paclitaxel +Cisplatin Cervical cancer In vivo Passive [173,237
PEG–PLL–PLLeu
poly(ethylene glycol)- b-poly(L-lysine)-b- poly(L-leucine) 
Docetaxel + Bcl-2 siRNA Breast cancer In vivo Passive [173,238
PCL-b-P(OEGMA-co- AzPMA
POEGMA- poly(OEGMA);
OEGMA- oligo(ethylene glycol) ethyl methacrylate;
co- copolymer;
AzPMA- 3-azidopropyl methacrylate 
Doxorubicin+ platinum(IV) Cervical cancer and melanoma In vitro Passive [173,239
DA3 (deoxycholic acid-conjugated PEI) Paclitaxel + XIAP siRNA Colorectal cancer In vivo Passive [173,240
Self-assembled polymeric micelles Paclitaxel + survivin siRNA Ovarian cancer In vivo Passive [173,241
P–H/M (methoxy poly(ethylene glycol)– poly(caprolactone) micelles) Paclitaxel + Honokiol Breast cancer In vivo Passive [173,242
Crosslinked PEG-b-pAsp-b-pTyr Docetaxel +lonidamine Breast cancer In vivo Passive [173,243
Water-soluble polymer conjugate-based co-delivery of drugs against cancer 
HPMA copolymer Doxorubicin+ dexamethasone General cancer In vivo Passive [18,244
HPMA copolymer TNP-470 + Alendronate Bone metastasis In vivo Active; bone [18,245
HPMA copolymer Paclitaxel + Alendronate Bone metastasis In vivo Active; bone [18,118
Branched PEG Epirubicin + Nitric oxide Colon cancer In vivo Passive [18,246–248
Branched PEG Camptothecin + BH3 domain peptide Ovarian primary tumor and metastatic malignant ascites In vivo Active; luteinizing hormone-releasing hormone [18,249
HPMA copolymer Trastuzumab + PKI166 HER2 overexpressed breast cancer In vitro Active; HER2 [18,250
HPMA copolymer 6.4 wt% gemcitabine + 5.7 wt% of Doxorubicin + 1.0 mol% tyrosinamide Prostate cancer In vivo Passive [173,251
Microsphere-based co-delivery of drugs against cancer 
Double-walled microspheres,PLGA core surrounded by PLLA shell (PLLA- poly(L-lactic acid)) Doxorubicin+ Chitosan p53 DNA Hepatocellular carcinoma In vitro Passive [173,252
Carbon nanoparticle and carbon-based nanosystem based co-delivery of drugs against cancer 
Nanodiamond Paclitaxel + Cetuximab Colorectal cancer In vivo Active, epidermal growth factor receptor positive cells [173,253
PEGylated lipid bilayer-wrapped nano-graphene oxide (GOLDR) Doxorubicin + Rapamycin Breast cancer In vitro Passive [173,254
Metallic nanoparticle-based co-delivery of drugs against cancer 
Silver nanoparticles (SN-AK-DOX) (SNs- silver nanoparticles; AK- sanazole) Sanazole + Doxorubicin Lymphoma In vivo Active, hypoxic cells [173,255
Gold nanoparticles Doxorubicin+ Cisplatin, +Capecitabine Hepatocellular carcinoma In vitro Passive [173,256
Carrier compositionTherapeuticsIndicationStatusTargetingRef.
Liposome based co-delivery of drugs against cancer 
PEG-Liposome Topotecan + Vincristine Brain cancer In vivo Passive [18,211
Polymer-caged nanobins (PCN); liposome surrounded by cholesterol-terminated poly(acrylic acid) Cisplatin + Doxorubicin Various cancers In vitro Passive [18
Liposome Cytarabine + Daunorubicin Acute myeloid leukemia Phase II Passive [18,212
Liposome Irinotecan + Floxuridine Colorectal cancer Phase II Passive [18,213,214
The mixture of two Liposomes Irinotecan + Cisplatin Small-cell lung cancer In vivo Passive [18,214
PEG-Liposome Quercetin + Vincristine Hormone- and TRZ insensitive breast cancer In vivo Passive [18,215
Cationic, anionic PEG Liposome VEGF/ c-myc siRNA + Doxorubicin MDR-breast cancer In vivo Passive [18,216
Liposome 6-Mercaptopurine + Daunorubicin Acute myeloid leukemia In vitro Passive [18,217
Dendrimer based co-delivery of drugs against cancer 
G5 PAMAM dendrimer (G5-Generation 5, PAMAM-poly(amidoamine)) Antisense-miRNA21 +5-fluorouracil Glioblastoma In vitro Active; miRNA overexpression [18,218
Aptamer-G4 PAMAM dendrimer conjugates (G4-Generation 4) Unmethylated CpG-oligonucleotides+ Doxorubicin Prostate cancer In vivo Active; a single-strand DNA-A9 prostate-specific membrane antigen, RNA aptamer hybrid [18,219
Dendritic PEG H2N–PEG–dendrimer– (COOH)4 Paclitaxel + alendronate Cancer bone metastasis In vivo Both passive and active Active by alendronate molecule [18,220
RGDfK-G3 Poly-lysine dendrimer (G3-Generation 3) Doxorubicin + siRNA Glioblastoma In vitro Active; αvβ3 integrin [18,221
Folate-G5 poly -propyleneimine dendrimer with ethylenediamine core (G5-Generation 5) Methotrexate + all-trans-retinoic acid Leukemia In vitro Active; folate receptor [18,222
Polymer based co-delivery of drugs against cancer 
PEG-PLGA Lonidamine + Paclitaxel Multiple drug resistant (MDR) breast cancer In vitro Active; EGFR [18,223
Methoxy PEG-PLGA Doxorubicin+ paclitaxel Various cancers In vitro Passive [18,223
PEG-PLA (PLA-poly(d,l, lactic acid)) Paclitaxel, Etoposide, or Docetaxel + 17-AAG Various cancers In vitro Active; HSP90 [18,224
PEG-PLA Combretastatin A4 + Doxorubicin Various cancers In vitro Active; angiogenesis [18,225
PDMAEMA-PCL- PDMAEMA poly(N,N-dimethylamino-2- ethyl methacrylate)- polycaprolactone-poly (N,N-dimethylamino- 2-ethyl methacrylate) Paclitaxel + VEGF siRNA Prostate cancer In vitro Active; VEGF [18,226
PEG-DSPE/PLGA Combretastatin + Doxorubicin Lung carcinoma In vitro Passive [18,115
PEG-PLA and PEG-DSPE/TPGS (TPGS- tocopheryl polyethylene glycol) Paclitaxel + 17-AAG(HSP90 inhibitor) Ovarian cancer In vitro Active; HSP90 [18,227
P(MDS-co-CES) poly (N-methyldietheneamine sebacate)- co-[(cholesteryl oxocarbonylamido ethyl) methyl bis(ethylene) ammonium bromide] Paclitaxel + Interleukin-12 or Bcl-2 siRNA Breast cancer In vivo Active; Bcl-2 [18,228
PEG-b-PHSA PEG-block-poly(N-hexyl stearate l-aspartamide) Doxorubicin + Wortmannin Breast cancer In vitro Passive [18,229
PLN formulation ((DG)n) (Polymer lipid hybrid nanoparticles (NP) co-loaded with DOX and GG918) Doxorubicin + GG918 Breast cancer In vitro Passive [230
PLGA Vincristine + Verapamil Hepatocellular carcinoma In vitro Passive [18,231
PLGA Paclitaxel + Tariquidar Breast cancer In vivo Active; functionalized with biotin [173,232
PLGA Rapamycin + piperine Breast cancer In vitro Passive [173,233
PACA polyalkylcyanoacrylate Doxorubicin + Cyclosporine A Various cancers In vitro Passive [18,234
PEG outer shell, middle PCL and inner CPCL core (PCL- polycaprolactone; CPCL- carboxylic functionalized PCL) Doxorubicin +Cisplatin Breast cancer In vitro Passive [173,235
Bradykinin-potentiating peptide decorated chitosan nanoparticle Bradykinin-potentiating peptide + bioreductively sensitive platinum (IV) compound which becomes cisplatin in intracellular reductive environment Hepatocellular carcinoma In vivo Passive [188,236
Polymeric micelles based co-delivery of drugs against cancer 
MPEG-b-P(LA-co-MCC) (MPEG-b-P(LA-co-MCC)) - methoxy poly(ethylene glycol)-block-poly(1-lactide -co-2-methyl-2- carboxyl-propylene carbonate) Paclitaxel +Cisplatin Cervical cancer In vivo Passive [173,237
PEG–PLL–PLLeu
poly(ethylene glycol)- b-poly(L-lysine)-b- poly(L-leucine) 
Docetaxel + Bcl-2 siRNA Breast cancer In vivo Passive [173,238
PCL-b-P(OEGMA-co- AzPMA
POEGMA- poly(OEGMA);
OEGMA- oligo(ethylene glycol) ethyl methacrylate;
co- copolymer;
AzPMA- 3-azidopropyl methacrylate 
Doxorubicin+ platinum(IV) Cervical cancer and melanoma In vitro Passive [173,239
DA3 (deoxycholic acid-conjugated PEI) Paclitaxel + XIAP siRNA Colorectal cancer In vivo Passive [173,240
Self-assembled polymeric micelles Paclitaxel + survivin siRNA Ovarian cancer In vivo Passive [173,241
P–H/M (methoxy poly(ethylene glycol)– poly(caprolactone) micelles) Paclitaxel + Honokiol Breast cancer In vivo Passive [173,242
Crosslinked PEG-b-pAsp-b-pTyr Docetaxel +lonidamine Breast cancer In vivo Passive [173,243
Water-soluble polymer conjugate-based co-delivery of drugs against cancer 
HPMA copolymer Doxorubicin+ dexamethasone General cancer In vivo Passive [18,244
HPMA copolymer TNP-470 + Alendronate Bone metastasis In vivo Active; bone [18,245
HPMA copolymer Paclitaxel + Alendronate Bone metastasis In vivo Active; bone [18,118
Branched PEG Epirubicin + Nitric oxide Colon cancer In vivo Passive [18,246–248
Branched PEG Camptothecin + BH3 domain peptide Ovarian primary tumor and metastatic malignant ascites In vivo Active; luteinizing hormone-releasing hormone [18,249
HPMA copolymer Trastuzumab + PKI166 HER2 overexpressed breast cancer In vitro Active; HER2 [18,250
HPMA copolymer 6.4 wt% gemcitabine + 5.7 wt% of Doxorubicin + 1.0 mol% tyrosinamide Prostate cancer In vivo Passive [173,251
Microsphere-based co-delivery of drugs against cancer 
Double-walled microspheres,PLGA core surrounded by PLLA shell (PLLA- poly(L-lactic acid)) Doxorubicin+ Chitosan p53 DNA Hepatocellular carcinoma In vitro Passive [173,252
Carbon nanoparticle and carbon-based nanosystem based co-delivery of drugs against cancer 
Nanodiamond Paclitaxel + Cetuximab Colorectal cancer In vivo Active, epidermal growth factor receptor positive cells [173,253
PEGylated lipid bilayer-wrapped nano-graphene oxide (GOLDR) Doxorubicin + Rapamycin Breast cancer In vitro Passive [173,254
Metallic nanoparticle-based co-delivery of drugs against cancer 
Silver nanoparticles (SN-AK-DOX) (SNs- silver nanoparticles; AK- sanazole) Sanazole + Doxorubicin Lymphoma In vivo Active, hypoxic cells [173,255
Gold nanoparticles Doxorubicin+ Cisplatin, +Capecitabine Hepatocellular carcinoma In vitro Passive [173,256

In some instances, co-delivery of drugs in a single nanoparticle suffers from drug leakage and poor loading efficacy [259]. To overcome this problem, researchers have conjugated two anticancer drugs via suitable covalent linkages and then subsequently encapsulated the conjugate in a nanoparticle. For example, Aryal et al. have used ester linkage for conjugating paclitaxel and gemcitabine and loaded the drug conjugate into a PLGA nanoparticle. Hydrolysis of this ester linkage at mildly acidic pH (pH 6) of endosomal environment resulted in two separate functional drug fragments [260]. Matlapudi et al. have conjugated Imatinib mesylate (abbreviated as IM, a tyrosine kinase inhibitor) and 5-fluorouracil (abbreviated as FU, an antimetabolite) by hydrolysable succinyl linker (abbreviated as Su) which forms amide linkage with each drug. This drug conjugate IM-Su-FU was incorporated in a human serum albumin (HSA) nanoparticle. This HSA encapsulated drug conjugate (IM-Su-FU) nanoparticle exhibited higher anticancer efficacy in in vivo lung cancer model compared to free drugs and only IM-Su-FU conjugate. Pharmacokinetic analysis of this nanoparticle exhibited improved elimination rate, half-life and mean residence time (MRT) than free drug and only IM-Su-FU conjugate [261].

Nanoparticle-mediated combination therapy has shown great potential in treating metastatic and drug resistant cancer [262]. Combination therapies involving cytotoxic drugs, signal transduction inhibitors, immunotherapeutic drugs, epigenetic agents and priming with apoptotic drugs have shown promising possibilities for cancer therapeutics [258]. The success of nanoparticle mediated combination owes to its “3R” delivery principle, i.e., right place, right dose and right time. To gain the benefits of nanocarrier mediated combination therapy, different databases and drug development platforms are developed. EMBASE® and Ovid MEDLINE® are databases used for the co-delivery of drugs. The drug combination development platform CombiPlex screens dual drugs and makes nanoscale formulation for nanocarrier mediated co-delivery of drugs. CombiPlex platform first determines the synergistic ratio of free drugs and then chooses a suitable nanocarrier to coordinate the pharmacokinetics of the free drugs and ensures the drugs reach the tumor site in the desired ratiometric manner. CPX-351, developed from CombiPlex platform is an FDA approved drug used for treating AML in adults. CPX-351 comprises cytarabine and daunorubicin in a synergistic ratio encapsulated in a liposomal carrier [257].

Although nanoparticle-mediated combination therapy has immense potential, but the regime of nanoparticle-based combination therapy is far from being able to cure metastatic cancers. 0.7% is the median efficiency of delivery of the injected nanoparticle to the desired tumor site [24]. Additionally, the five-year survival rates of most malignant cancer are still quite low and most combination approaches still depend on cytotoxic approaches instead of molecularly targeted anticancer agents or nanoparticle mediated combination approach [263]. Saptura et al. has shown antagonistic drugs interactions used in combination can suppress the clonal expression of singly-resistant cells in in silico model [264]. So antagonistic drug interactions should also be investigated and not completely discarded. Although around five thousand clinical trials are ongoing worldwide for the development of new combination therapy, Palmer and Sorger, 2017 claims that most of the combination therapy used follows the independent action model and provide therapeutic benefit due to patient-to-patient variability rather than additive or synergistic drug interactions [265,266].

One of the key challenges in developing nanoparticle-based targeted therapy is the issue of toxicity of treatment combinations. Seemingly rationally developed and assumed to be safe combination therapy in preclinical model may fail in clinical trials due to toxicity, especially in the case of synergistic drugs which can lead to synergistic toxicity as well due to similar mechanism of action or due to auto immune response of healthy tissue [169]. Mathematical modeling has been helpful in predicting toxicity in pre-clinical trials and can be used as an efficient tool for predicting the toxicity of drug combinations in a patient-specific manner [169]. Toxicity of the drug nanocarrier also needs to be considered. Another hurdle in the success of combinatorial nanotherapeutics to be translated into clinics is the lack of patients who are interested in participating in clinical trials. Approximately 40% of cancer trials fail due to the scarcity of patients [169]. Cancer is a highly heterogeneous disease and cell type population in a particular type of malignant tumor varies from patient to patient. For clinical trials to have higher success rates, efficient biomarkers need to be identified first. Additionally, clinical trials should be performed on patients with a similar level of biomarker expression. But due to the lack of patients enrolled in a clinical trial, such arrangements are not possible. Due to the insufficiency of suitable patients for performing clinical trials, precise preclinical models need to be developed. Researchers need to look beyond immune-competent mice model and use patient-derived xenograft in vivo models, humanized mouse models and patient-derived organoids for a greater chance of success in clinical trials and such models can also be used as an efficient tool for predicting the toxicity of drug combinations in a patient specific manner [169].

Immunotherapy is a monumental breakthrough recently included in the existing therapeutic armamentarium against cancer. It utilizes body’s own immune system to fight cancer. William Coley, known as the father of immunotherapy, first attempted to treat cancer utilizing the immune system. However, this field got attention when James P. Allison and Tasuku Honjo got the Nobel prize for cancer immunotherapy in the year 2018. Judy Perkins having stage IV metastatic breast cancer, was the first lady to be cured successfully by immunotherapy [267]. Current cancer immunotherapy comprises cytokine therapy, antibody-based therapy and adoptive cell therapy. However, complex tumor microenvironment limits the efficacy of immunotherapy. In the tumor microenvironment, tumor cells can polarize tumor associated macrophages (TAM) toward pro-tumorigenic M2 macrophages while decreasing antitumorigenic M1 macrophages to facilitate tumor progression. Moreover, cancer cells help to activate immune checkpoints leaving the T cells in a state of anergy (lack of responsiveness to an antigen). Cancer immunotherapy inhibits immune checkpoints to overcome the T-cell anergy, thereby its activation against cancer. Ipilimumab inhibits the cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4) checkpoint in T cells leading to its activation. Similarly, programmed cell death protein 1 (PD-1) in T cells is inhibited by FDA-approved anti PD-1 antibody pembrolizumab and nivolumab. Cancer cells overexpress the immune checkpoint protein programmed death-ligand 1 (PD-L1). The PD-L1 inhibiting antibodies atezolizumab, avelumab, durvalumab are also approved by FDA [268]. These antibodies block the inhibitory signal induced by interaction of PD-1 of T cell to PD-L1 of the cancer cell. Multiple colony stimulating factor receptor 1 (CSF-1R) inhibitors are currently in clinical trials for their ability to polarize tumor associated macrophages to anti-tumorigenic M1 phenotype [13].

Peptide-based drugs/vaccines can hugely advance cancer immunotherapy. Peptides can be designed in a more rationalized manner to target desired molecule of tumor or the tumor microenvironment. Yin et al. have designed a peptide (IQIREYKRCGQDEERVRRECKERGERQNCHYVIHKEGNCYVCGIICL) mimicking the native structure of the PD-1 molecule, which inhibited the interaction between PD-L1 of cancer cells and PD-1 of immune cells. This peptide was developed as a potent drug for cancer immunotherapy [269]. Hazama et al. also developed a peptide-based macrocyclic peptide (c[Ac-DYRYSAVYSIHPSWC]G) inhibiting the interaction between cluster of differentiation 47 (CD47) of cancer cell and signal regulatory protein α (SIRPα) of macrophages and showed its in vivo efficacy [270]. Peptide-based cancer vaccines can specifically stimulate cancer specific T cell response [271]. Peptide-based (9 amino acid residues) vaccine GP2 (IISAVVGIL) is currently in phase 2 clinical trial against breast cancer [272]. Several other examples of peptide vaccines combined with other drugs are highlighted in Table 10.

Researchers have reported a number of combinatorial approaches where immunotherapeutic drugs are combined with the conventional cancer therapeutics such as chemotherapy, RNAi therapy, photothermal, photodynamic and radiotherapy as described in Tables 12-14 and Figure 11.

Next-generation combination nanomedicine for immunotherapy

Figure 11
Next-generation combination nanomedicine for immunotherapy

(A) CD47 (cluster of differentiation 47; a transmembrane protein) is overexpressed on cancer cells and binds to SIRPα (signal regulatory protein α; a regulatory membrane glycoprotein) on immune cells to inactivate immune cells and escape immune surveillance. Similarly, MCSF (macrophage colony stimulating factor; a secretory protein) released from cancer cells which binds to CSF-1R (colony stimulating factor 1 receptor; a transmembrane protein) on immune cells and inactivates immune cells. A supramolecular nanoparticle, comprising anti-SIRPα antibody with a small molecule inhibitor (BLZ-945) of CSF-1R inhibits both the signaling axes simultaneously and shows anti-tumor immune response. (B) Nanoparticle having immune checkpoint inhibitor and a small molecule-based kinase inhibitor causes targeted disruption of kinase signaling only in cancer cells while keeping the PI3K and MAPK pathways of immune cells untouched. This strategy enables inhibition of kinase signaling. (C) Chemokine CXCL12 (C-X-C motif chemokine ligand 12), secreted from fibroblast cells binds to CXCR4 (C-X-C chemokine receptor 4), a G-protein-coupled receptor from cancer cells and promotes immunosuppressive tumor microenvironment. Combination immunotherapy with inhibitors of CXCR4/CXCL12 axis can amplify the antitumor efficacy. (D) combination of epigenetic inhibitor (Zebularine, a DNMTi) with immune checkpoint inhibitor of PD-L1 can potentiate combination immunotherapy.

Figure 11
Next-generation combination nanomedicine for immunotherapy

(A) CD47 (cluster of differentiation 47; a transmembrane protein) is overexpressed on cancer cells and binds to SIRPα (signal regulatory protein α; a regulatory membrane glycoprotein) on immune cells to inactivate immune cells and escape immune surveillance. Similarly, MCSF (macrophage colony stimulating factor; a secretory protein) released from cancer cells which binds to CSF-1R (colony stimulating factor 1 receptor; a transmembrane protein) on immune cells and inactivates immune cells. A supramolecular nanoparticle, comprising anti-SIRPα antibody with a small molecule inhibitor (BLZ-945) of CSF-1R inhibits both the signaling axes simultaneously and shows anti-tumor immune response. (B) Nanoparticle having immune checkpoint inhibitor and a small molecule-based kinase inhibitor causes targeted disruption of kinase signaling only in cancer cells while keeping the PI3K and MAPK pathways of immune cells untouched. This strategy enables inhibition of kinase signaling. (C) Chemokine CXCL12 (C-X-C motif chemokine ligand 12), secreted from fibroblast cells binds to CXCR4 (C-X-C chemokine receptor 4), a G-protein-coupled receptor from cancer cells and promotes immunosuppressive tumor microenvironment. Combination immunotherapy with inhibitors of CXCR4/CXCL12 axis can amplify the antitumor efficacy. (D) combination of epigenetic inhibitor (Zebularine, a DNMTi) with immune checkpoint inhibitor of PD-L1 can potentiate combination immunotherapy.

Close modal
Table 12
Combination nanomedicine having immunotherapeutic drugs with chemotherapeutic drugs
Chemotherapeutic drugsImmunotherapeutic drugsNanoparticle delivery systemCancer TypeOutcomeRef.
Doxorubicin Anti–PD-1 antibody Synthetic high-density lipoprotein (sHDL) like nanodiscs loaded with DOX CT26 and MC38 mouse colon carcinoma Induced strong anticancer immunity and sensitized tumors to immune checkpoint blockade [273
Doxorubicin Cytosine–phosphate–guanosine oligonucleotides (CpG-ONT) (Immune-stimulating agent) An RNA aptamer (recognizing a prostate-specific membrane antigen (PSMA)) bioconjugated with a dendrimer attached with CpG-ONTs loaded with DOX In vivo and in vitro models of prostate cancer (22RV1) Showed excellent antitumor efficacy, immune stimulation and target specificity [219
Paclitaxel Toll-like receptor-7 (TLR-7) agonist-imiquimod Poly (γ-glutamic acid) (γ-PGA) micro-dispersion system of drugs In vitro and in vivo mouse melanoma (B16-F10) Showed robust immunogenic tumor cell death followed by inhibition of secondary tumors also [274
Paclitaxel TLR-4 agonist bacterial endotoxin Lipopolysaccharide (LPS) Co-encapsulation by PLGA-based nanoparticle In vitro and in vivo murine melanoma model (B16-F10) Tumor volume was found 40% less and immune activation was observed [275
Paclitaxel Cytosine–phosphate–guanosine oligodeoxynucleotides (CpG ODNs) and IL-10 siRNA (Immune-stimulating agents) PLGA-based nanoparticles Murine melanoma model (B16-F10) Efficiently inhibited tumor growth and increased the animal survival rate [276
Cisplatin Cytosine–phosphate–guanosine (CpG) (Immune-stimulating agent) Liposome Murine melanoma model (B16-F10) Strong synergistic effect which increased apoptosis and reduced tumor growth [277
Mitoxantrone treated CT26 cancer cells decorated with Cytosine– phosphate–guanosine (CpG) (a potent TLR9 agonist) loaded nanoparticle Anti-PD-1 antibody Hyaluronic acid-cationic lipid nanoparticle was loaded with CpG Murine model of melanoma and colon carcinoma (B16-F10-OVA and CT26) Complete tumor regression in almost 78% of CT26 tumor-bearing mice and long-term immunity against tumor recurrence [278
Doxorubicin and immune stimulating agent CpG-loaded microparticles Immune checkpoint inhibitor antibodies anti-CTLA-4 and anti-OX40 PLGA-based microparticles Mouse lymphoma (EL4, E20) and mouse melanoma (B16-fLUC) models Generated systemic immune responses that suppressed injected and distant tumors in a murine B lymphoma model, leading to tumor-free mice Reduced tumor burdens [279
Doxorubicin Immunotherapeutic agent interferon-γ (IFN-γ) PLGA-based thermosensitive nanoparticle Murine melanoma model (B16-F10) Prolonged circulation time, sustained drug release, excellent synergistic antitumor efficiency against B16F10 tumor bearing mice [280
Paclitaxel Interleukin-2 Hydroxypropyl-β- cyclodextrin acrylate and two opposite charged chitosan derivatives based nanogels coated by RBC membrane Murine melanoma model (B16-F10) Enhanced antitumor activity with improved drug penetration and increased antitumor immunity [281
Doxorubicin (DOX), all-trans retinoic acid (ATRA), Interleukin-2 Lipid-coated biodegradable hollow mesoporous silica nanoparticle (dHMLB) Murine melanoma model (B16-F10) Significant tumor growth and metastasis inhibition [282
Chemotherapeutic drugsImmunotherapeutic drugsNanoparticle delivery systemCancer TypeOutcomeRef.
Doxorubicin Anti–PD-1 antibody Synthetic high-density lipoprotein (sHDL) like nanodiscs loaded with DOX CT26 and MC38 mouse colon carcinoma Induced strong anticancer immunity and sensitized tumors to immune checkpoint blockade [273
Doxorubicin Cytosine–phosphate–guanosine oligonucleotides (CpG-ONT) (Immune-stimulating agent) An RNA aptamer (recognizing a prostate-specific membrane antigen (PSMA)) bioconjugated with a dendrimer attached with CpG-ONTs loaded with DOX In vivo and in vitro models of prostate cancer (22RV1) Showed excellent antitumor efficacy, immune stimulation and target specificity [219
Paclitaxel Toll-like receptor-7 (TLR-7) agonist-imiquimod Poly (γ-glutamic acid) (γ-PGA) micro-dispersion system of drugs In vitro and in vivo mouse melanoma (B16-F10) Showed robust immunogenic tumor cell death followed by inhibition of secondary tumors also [274
Paclitaxel TLR-4 agonist bacterial endotoxin Lipopolysaccharide (LPS) Co-encapsulation by PLGA-based nanoparticle In vitro and in vivo murine melanoma model (B16-F10) Tumor volume was found 40% less and immune activation was observed [275
Paclitaxel Cytosine–phosphate–guanosine oligodeoxynucleotides (CpG ODNs) and IL-10 siRNA (Immune-stimulating agents) PLGA-based nanoparticles Murine melanoma model (B16-F10) Efficiently inhibited tumor growth and increased the animal survival rate [276
Cisplatin Cytosine–phosphate–guanosine (CpG) (Immune-stimulating agent) Liposome Murine melanoma model (B16-F10) Strong synergistic effect which increased apoptosis and reduced tumor growth [277
Mitoxantrone treated CT26 cancer cells decorated with Cytosine– phosphate–guanosine (CpG) (a potent TLR9 agonist) loaded nanoparticle Anti-PD-1 antibody Hyaluronic acid-cationic lipid nanoparticle was loaded with CpG Murine model of melanoma and colon carcinoma (B16-F10-OVA and CT26) Complete tumor regression in almost 78% of CT26 tumor-bearing mice and long-term immunity against tumor recurrence [278
Doxorubicin and immune stimulating agent CpG-loaded microparticles Immune checkpoint inhibitor antibodies anti-CTLA-4 and anti-OX40 PLGA-based microparticles Mouse lymphoma (EL4, E20) and mouse melanoma (B16-fLUC) models Generated systemic immune responses that suppressed injected and distant tumors in a murine B lymphoma model, leading to tumor-free mice Reduced tumor burdens [279
Doxorubicin Immunotherapeutic agent interferon-γ (IFN-γ) PLGA-based thermosensitive nanoparticle Murine melanoma model (B16-F10) Prolonged circulation time, sustained drug release, excellent synergistic antitumor efficiency against B16F10 tumor bearing mice [280
Paclitaxel Interleukin-2 Hydroxypropyl-β- cyclodextrin acrylate and two opposite charged chitosan derivatives based nanogels coated by RBC membrane Murine melanoma model (B16-F10) Enhanced antitumor activity with improved drug penetration and increased antitumor immunity [281
Doxorubicin (DOX), all-trans retinoic acid (ATRA), Interleukin-2 Lipid-coated biodegradable hollow mesoporous silica nanoparticle (dHMLB) Murine melanoma model (B16-F10) Significant tumor growth and metastasis inhibition [282
Table 13
Combination nanomedicine having immunotherapeutic drugs with siRNA
Gene therapyImmunotherapeutic drugsNanoparticle delivery systemCancerOutcomeRef.
Signal transducer and activator of transcription-3 (STAT3) silencing siRNA TLR-7 agonist imiquimod, R837 (immune response modifier) PLGA NPs Murine T-cell lymphoma model (EG7-OVA) Inhibited tumor growth efficiently [283
IL10-silencing siRNA CpG ODN Cationic PLGA-PEI microparticles Murine model of B cell lymphoma (A20) Better immune protection of an idiotype DNA vaccine [284
TGF-β silencing siRNA A mannose-modified lipid-calcium-phosphate Nanoparticle based vaccine containing tumor antigen (Trp 2 peptide) and adjuvant (CpG oligonucleotide) Liposome-protamine- hyaluronic acid (LPH) NP Murine melanoma model (B16F10) Boosted the vaccine efficacy and inhibited tumor growth by 52% [285
IL-6 silencing siRNA Radiofrequency thermal ablation Micelle like nanoparticle Mouse breast adenocarcinoma (R3230 and MATBIII) Reduced tumor growth [286
PD-L1 silencing siRNA Photodynamic therapy Acid-activatable cationic micelle Murine melanoma model (B16F10) Significantly enhanced efficacy for inhibiting tumor growth and distant metastasis [287
Gene therapyImmunotherapeutic drugsNanoparticle delivery systemCancerOutcomeRef.
Signal transducer and activator of transcription-3 (STAT3) silencing siRNA TLR-7 agonist imiquimod, R837 (immune response modifier) PLGA NPs Murine T-cell lymphoma model (EG7-OVA) Inhibited tumor growth efficiently [283
IL10-silencing siRNA CpG ODN Cationic PLGA-PEI microparticles Murine model of B cell lymphoma (A20) Better immune protection of an idiotype DNA vaccine [284
TGF-β silencing siRNA A mannose-modified lipid-calcium-phosphate Nanoparticle based vaccine containing tumor antigen (Trp 2 peptide) and adjuvant (CpG oligonucleotide) Liposome-protamine- hyaluronic acid (LPH) NP Murine melanoma model (B16F10) Boosted the vaccine efficacy and inhibited tumor growth by 52% [285
IL-6 silencing siRNA Radiofrequency thermal ablation Micelle like nanoparticle Mouse breast adenocarcinoma (R3230 and MATBIII) Reduced tumor growth [286
PD-L1 silencing siRNA Photodynamic therapy Acid-activatable cationic micelle Murine melanoma model (B16F10) Significantly enhanced efficacy for inhibiting tumor growth and distant metastasis [287
Table 14
Combination nanomedicine of immunotherapeutic drugs with photothermal, photodynamic and radiotherapy
Photothermal/ photodynamic/ radiotherapyImmunotherapeutic drugsNanoparticle delivery systemCancerOutcomeRef.
Combination of immunotherapeutic drugs with photothermal therapy 
Photothermal ablation (near infrared light) Immunoadjuvants oligodeoxynucleotides containing the cytosineguanine (CpG) motifs Chitosan coated hollow Copper Sulfide nanoparticles Murine breast cancer model (EMT6-OVA, EMT6) Combined photothermal immunotherapy is more effective than immunotherapy/ photothermal therapy alone in mouse breast cancer model [288
Photothermal ablation (near infrared laser) Adoptive T cell therapy Gold nanoshell Murine melanoma (B16-F10) Prevents primary tumor recurrence post-ablation, inhibited tumor growth at distant sites, and abrogated the outgrowth of lung metastases [289
Photothermal ablation Anti-CTLA-4 antibody Single-walled carbon nanotube Murine model of breast cancer (4T1) Tumor metastasis prevented [290
Photothermal ablation Gold nanostar Anti-PDL1 antibody Mouse bladder cancer (MB49) Both primary and distant tumors were safely eradicated [291
Combination immunotherapeutic drugs with photodynamic therapy 
Photosensitizer pyropheophorbide-lipid conjugate (pyrolipid) in the shell and oxaliplatin in the core Anti PD-L1 antibody Nanoscale coordination polymer (NCP) core-shell nanoparticles Murine colorectal tumor (CT26 and MC38) This combination causes regression of both primary and distant tumors via induction of strong cancer specific immune response [292
Photosensitizer pyrolipid (ZnP@pyro) Anti PD-L1 antibody Zn-pyrophosphate (ZnP) nanoparticles Murine breast cancer model (4T1) Complete eradication of primary and distant tumors via systemic cancer specific cytotoxic T-cell response [293
Chlorin e6 (Ce6), a photosensitizer and imiquimod (R837), a Toll-like-receptor-7 agonist Anti-CTLA-4 antibody Upconversion nanoparticles (UCNPs) Murine colon carcinoma (CT26) Eliminates NIR laser exposed tumors but causes strong anticancer immunity to inhibit distant tumors also [294
TBC-Hf (derived fromn tetra(pbenzoato)chlorin and Hf) enabled photodynamic therapy Small-molecule inhibitor of indoleamine 2,3-dioxygenase (IDO) Chlorin-based nanoscale metal−organic framework (nMOF) Murine colorectal models (CT26 and MC38) Effective local and distant tumor rejection in colorectal cancer models [295
Combination immunotherapeutic drugs with radiotherapy 
Radiation therapy Cowpea-mosaic virus Cowpea-mosaic virus nanoparticle Murine ovarian cancer (ID8- Defb29/Vegf-A-Luc cells) Resulted in improved tumor growth delay and an increase in tumor infiltrating lymphocytes (TILs) [296
Radiotherapy Small molecule based indoleamine 2,3-dioxygenase (IDO) inhibitor Hafnium (Hf)-based nanoparticle Mouse models of breast and colorectal cancer Eradication of local and distal tumors in in vivo models [297
Photothermal/ photodynamic/ radiotherapyImmunotherapeutic drugsNanoparticle delivery systemCancerOutcomeRef.
Combination of immunotherapeutic drugs with photothermal therapy 
Photothermal ablation (near infrared light) Immunoadjuvants oligodeoxynucleotides containing the cytosineguanine (CpG) motifs Chitosan coated hollow Copper Sulfide nanoparticles Murine breast cancer model (EMT6-OVA, EMT6) Combined photothermal immunotherapy is more effective than immunotherapy/ photothermal therapy alone in mouse breast cancer model [288
Photothermal ablation (near infrared laser) Adoptive T cell therapy Gold nanoshell Murine melanoma (B16-F10) Prevents primary tumor recurrence post-ablation, inhibited tumor growth at distant sites, and abrogated the outgrowth of lung metastases [289
Photothermal ablation Anti-CTLA-4 antibody Single-walled carbon nanotube Murine model of breast cancer (4T1) Tumor metastasis prevented [290
Photothermal ablation Gold nanostar Anti-PDL1 antibody Mouse bladder cancer (MB49) Both primary and distant tumors were safely eradicated [291
Combination immunotherapeutic drugs with photodynamic therapy 
Photosensitizer pyropheophorbide-lipid conjugate (pyrolipid) in the shell and oxaliplatin in the core Anti PD-L1 antibody Nanoscale coordination polymer (NCP) core-shell nanoparticles Murine colorectal tumor (CT26 and MC38) This combination causes regression of both primary and distant tumors via induction of strong cancer specific immune response [292
Photosensitizer pyrolipid (ZnP@pyro) Anti PD-L1 antibody Zn-pyrophosphate (ZnP) nanoparticles Murine breast cancer model (4T1) Complete eradication of primary and distant tumors via systemic cancer specific cytotoxic T-cell response [293
Chlorin e6 (Ce6), a photosensitizer and imiquimod (R837), a Toll-like-receptor-7 agonist Anti-CTLA-4 antibody Upconversion nanoparticles (UCNPs) Murine colon carcinoma (CT26) Eliminates NIR laser exposed tumors but causes strong anticancer immunity to inhibit distant tumors also [294
TBC-Hf (derived fromn tetra(pbenzoato)chlorin and Hf) enabled photodynamic therapy Small-molecule inhibitor of indoleamine 2,3-dioxygenase (IDO) Chlorin-based nanoscale metal−organic framework (nMOF) Murine colorectal models (CT26 and MC38) Effective local and distant tumor rejection in colorectal cancer models [295
Combination immunotherapeutic drugs with radiotherapy 
Radiation therapy Cowpea-mosaic virus Cowpea-mosaic virus nanoparticle Murine ovarian cancer (ID8- Defb29/Vegf-A-Luc cells) Resulted in improved tumor growth delay and an increase in tumor infiltrating lymphocytes (TILs) [296
Radiotherapy Small molecule based indoleamine 2,3-dioxygenase (IDO) inhibitor Hafnium (Hf)-based nanoparticle Mouse models of breast and colorectal cancer Eradication of local and distal tumors in in vivo models [297

Combination chemotherapy with cancer immunotherapy is a wide area of interest. In phase III clinical trial, albumin nanoparticle bound chemotherapeutic drug paclitaxel (Abraxane®) combined with atezolizumab (Tecentriq®; FDA approved antibody of PD-L1) has recently demonstrated its efficacy in patients with advanced triple-negative breast cancer (TNBC) [22,298]. However, some patients showed grade 3 (moderate to severe) or grade 4 (life threatening symptoms) immune related adverse events [298]. Hence, combination of nanoparticle-mediated chemo-immunotherapy may facilitate targeted and localized delivery to increase the therapeutic index, reduce off-target toxicity and incidences of immune related adverse events. Tables 12 shows some examples of preclinical studies on combination of nanoparticle mediated chemotherapy and immunotherapy.

RNA interference (RNAi) therapy with siRNA, microRNA (miRNA) and short hairpin RNA (shRNA) is utilized to silence genes of specific signaling molecules, cytokines and chemokines. However, this therapy possesses certain limitations like rapid degradation of RNA-based drugs in circulation due to presence of nucleases, renal clearance, poor cellular uptake because of anionic nature, etc [299]. Therefore, nanoparticles have been used to circumvent such barriers. Gene therapy has been utilized in targeting immune checkpoints like PD-1-PD-L1 pathway [300]. Also, these can be used with other cancer treatment modalities to induce immune stimulation against cancer. Few examples are given in Table 13 where combination nanomedicine having immunotherapeutic drugs showed enhanced efficacy.

Photothermal, photodynamic and radiotherapies are also used in combination with immunotherapy. In photothermal therapy, cancer cells can be destroyed and eliminated from the tumor tissues at 40–44°C temperature due to DNA damage, protein denaturation and disruption of cellular membrane [20]. However, it requires high temperature for complete cell death, thus large tumors are prone to relapse. Thus combining photothermal therapy with immune-stimulating agents and nanoparticles can synergistically induce anti-tumor efficacy for the treatment of large established tumors and distant metastases.

Photodynamic therapy comprising combination of light with photosensitizers leads to the generation of reactive oxygen species (ROS) and subsequently damage of subcellular organelles [301]. However, as a monotherapy, it does not efficiently regress the tumor in an immunosuppressive tumor microenvironment [20]. It induces immunosuppression by the release of immunosuppressive cytokines. It provokes damage to the normal cells by releasing the self-antigens [302]. Thus, to balance the immunosuppressive effects of mono-photodynamic therapy, immunotherapy needs to be combined with it.

In radiotherapy, high energy ionizing radiation such as X-rays are utilized to cause free radical generation mediated DNA and cellular damage leading to cellular death [303]. However, radiation therapy helps to develop anticancer therapy, its solo use promotes immunosuppressive environment around tumor by recruiting immunosuppressive Treg cells [304]. It causes increased production of immune-inhibitory molecules such as PD-L1 and transforming growth factor-β (TGFβ) [305]. Thus, to overcome these problems of radio-monotherapy, combination of radiotherapy with immunotherapy is implicated. Some examples are given in Table 14.

Immune checkpoint inhibitors were initially used in combination with other conventional chemotherapeutic drugs or radiotherapy (Tables 12-14) to potentiate the tumor regression. At present, nanomedicines for immunotherapy combines mechanistically inspired drug which can demonstrate a synergistic effect. Sengupta and co-workers have designed a supramolecular bifunctional nanomedicine comprising amphiphiles which inhibits interaction of CD47 of cancer cell and SIRPα of macrophages and simultaneously the interaction of macrophage colony stimulating factor (MCSF) and colony stimulating factor 1 receptor (CSF-1R). This nanomedicine increases the M2 to M1 repolarization within the tumor microenvironment and improves the anticancer and antimetastatic efficacy in melanoma and breast cancer model [306]. Mitragotri and co-workers designed a class of phagocytosis-resistant discoidal particles containing interferon-γ (IFN-γ). These particles showed efficient adherence to macrophages and directed their polarization toward anti-tumor M1 phenotype in murine breast cancer model [307]. Other such combinations are described in the next section.

Next-generation cancer immunotherapy needs to be patient-specific to enhance its antitumor efficacy. Immunotherapeutic drugs can be combined with signaling pathway inhibitors for better synergistic outcomes and to amplify the efficacy of personalized medicine [308]. MEK inhibitors are used in combination with PD-L1 antibodies as MEK inhibition causes up-regulation of PD-L1 in cancer cells. In cancer, the dysregulated PI3K-AKT pathway also regulates the PD-L1 expression [309]. Kulkarni et al. designed a nanomedicine combining PD1-PDL1 immune checkpoint inhibitor with kinase (MEK and PI3K) inhibitors for enhanced anti-tumor efficacy [81]. It has also been reported that second generation anti-histaminic drugs like cetirizine, etc., in combination with immunotherapy, have improved the anticancer efficacy [310].

Combination of G-protein-coupled receptor (GPCR) inhibitors and immune checkpoint inhibitors can amplify antitumor efficacy. CXCR4, a GPCR, is overexpressed in cancer cells which mediates cell proliferation, tumor growth, metastasis and tumor relapse [311]. CXCL12, the chemokine binding to the CXCR4, is secreted from fibroblast cells in the tumor microenvironment. CXCL12/CXCR4 axis is another molecular target for cancer treatment as this axis leads to an immunosuppressive tumor microenvironment. Thus, combining plerixafore (AMD3100; CXCR4 inhibitor) with immune checkpoint inhibitor anti-PDL1 antibody showed decreased tumor volume [312].

Epigenetic drugs can potentiate combination immunotherapy [313]. DNA hypomethylating agent 5-aza-2′-deoxycytidine (5-AZA-CdR) (a DNA methyltransferase inhibitor, DNMTi) combined with an anti-CTLA-4 monoclonal antibody showed enhanced antitumor efficacy in murine model of breast cancer [314]. In Phase Ib clinical trial, the epigenetic drug guadecitabine was combined with ipilimumab (anti-CTLA-4 antibody) for patients having stage III/IV melanoma. The DNA hypomethylating drugs cause upregulation of genes CD274, PDCD1LG2, and CTLA-4 in the patients where resistance has developed. Currently, a PEG-based nanoparticle system was utilized to deliver plasmid-encoding shPD-L1 (this plasmid down-regulated expresion of PD-L1 protein of cancer cells) in combination with Zebularine (a DNMTi, causing overexpression of major histocompatibility complex I [MHC-I] expression). This combination can effectively initiate anticancer immunity and prevent tumor relapse by a strong anticancer memory [315].

Currently, immunotherapy is the latest approach that has emerged in finding a cure for cancer. However, only immunotherapy is insufficient to eradicate tumors due to : (1) lack of specificity and systemic toxicity, (2) low patient response rate, (3) variable immune contexture of patients, etc., (4) impaired immune function and antibody activity due to the acidic tumor microenvironment (Warburg effect), (5) divergent immune pattern of different organs, (6) development of resistance against immunotherapy, (7) autoimmune adverse effect generation by immune checkpoint blockade, (8) increased immune suppression with aging and (9) huge cost [316–318].

To advance the area of cancer immunotherapy in near future, toxicity issues associated with immunotherapy need to be addressed. Since cancer is a highly heterogeneous disease, nanocarrier encapsulated precision combinatorial immunotherapy will hold a great prospect in future cancer therapy. Tumor heterogeneity can be explored by the determination of biomarkers via genomic analysis of circulating cancer cells. Next-generation sequencing can be deployed to explore immunogenicity of mutated genes. These data along with resources like cBioPortal, Project Genie can be utilized in designing patient-specific immunotherapy in an effective and safer manner. Immunotherapy can be further boosted by utilizing combinatorial approaches. This would require identification of optimal dosing and delivering the same dose to the tumor site. This can be achieved by ratiometric dosing as described earlier. Mono anti-PD-1 therapy often causes compensatory activation of other checkpoints (e.g., CTLA-4), leading to immunosuppression [319]. Thus, simultaneous blocking of inter-related checkpoints is necessary to design effective combinatorial immunotherapy. The gut microbiome or blood supply to the tumor site can also alter responses to cancer immunotherapy by indirectly modifying tumor microenvironment [320]. Hence modulating these factors can help combating resistance to immunotherapy. Furthermore, antagonizing low pH of tumor microenvironment by acidity modulating drugs (proton-pump inhibitors) might act as a possible choice to overcome tumor resistance, potentiating the existing immunotherapy [318]. Thus, optimizing the current immunotherapy with personalized combinatorial nanomedicines can advance the development of next-generation cancer nano-immunotherapy.

The challenges of next-generation nanoparticle mediated multicomponent combination therapy and combination immunotherapy need to amplify the therapeutic potential by providing the enhanced stability of cancer drugs in biologically active form, reducing toxicity related issues, overcoming immunosuppression and preferentially accumulating at the tumor site. Due to lack of fenestered or discontinuous endothelium, effective delivery of clinically safe nanomedicine to less accessible tissues remains a considerable challenge. To achieve clinically potential, optimized nanoparticle designing, innovation in designing the mechanistically inspired nanoparticle, targeting specific metastatic foci, their delivery process and in vivo biodistribution based on structure and activity need detailed understanding. Integrating cancer biology and anti-metastatic nanotechnology, new strategies need to be engineered considering the biological mechanisms of various stages of metastasis. Tumor microenvironment specific targeted therapeutic intervention is desirable to improve the outcome. New guidelines need to be developed about engineering multicomponent combination therapy and it requires extensive studies on various classes of drug combinations, our understanding about inter-drug interactions for such cases, spatio-temporal release of anticancer drugs, nonspecific activation of the immune system by such multicomponent combination nanomedicine, diversity of metastatic foci, diversity of organ environment and delivery of biologically active multicomponent nanomedicine at the epicentre of solid tumor. Such therapeutics need to be developed by integrating clinical trials with an adequate number of patients having similar biomarker expression and the development of better pre-clinical models. Next-generation multifunctional cancer nanomedicine and nanoparticle mediated combination immunotherapy having mechanistically rational therapeutic combinations need to promote “multi-targeted therapy” by disrupting the adaptive chemoresistance and potentiating the effect of therapeutic combinations. Integrating proteomics data of patient samples, combination nanotherapy and immune-oncology, one may develop highly effective precision nanomedicine.

The authors declare the following competing financial interests: We have filed patents on peptide based siRNA transporters (application no: 201931001645, 201731040460) and peptide based cancer drugs (application no: 201731046166). R.S.R., Prof. Dhananjay Bhattacharyya (D.B.), A.M.M., Dr Abhijit Biswas (A.B.), K.C. and Dr Sanchita Mukherjee (S.M.) are inventors in the patent application no: 201931001645. R.S.R., D.B., Dr Chiranjit Dutta, A.B., K.C., S.M. and Paramita Gayen are inventors in the patent application no: 201731040460. R.S.R., D.B., C.D., K.C., A.B., P.G. and S.M. are inventors in the patent application no: 201731046166.

RSR thanks SERB-Power fellowship grant [grant number SPF/2021/000024]; DST SERB [grant number EMR/2016/003829]; and DBT Nanobiotechnology [grant number BT/PR22086/NNT/28/1222/2017] for funding.

Dr Kasturee Chakraborty, Archana Tripathi, Sukumar Mishra, Argha Mario Mallick and Dr Rituparna Sinha Roy designed and drafted this review article. All authors had read and approved the final manuscript.

R.S.R. thanks SERB-Power fellowship. K.C. and S.M. thank CSIR for fellowship, A.M.M. thanks CSIR and DBT Nanobiotechnology (BT/PR27059/NNT/28/1543/2017) for fellowship and A.T. thanks IISER Kolkata for Ph.D. fellowship.

CD47

cluster of differentiation 47

CSF-1R

colony stimulating factor 1 receptor

CTLA-4

cytotoxic T-lymphocyte–associated antigen 4

DNMTi

DNA methyltransferase inhibitor

GPCR

G-protein-coupled receptor

IFN-γ

interferon-γ

MCSF

macrophage colony stimulating factor

MHC-I

major histocompatibility complex I

NIR

near infrared

PD-1

programmed cell death protein 1

PD-L1

programmed death-ligand 1

SIRPα

signal regulatory protein α

TAM

tumor associated macrophages

TGFβ

transforming growth factor-β

1.
Sung
H.
,
Ferlay
J.
,
Siegel
R.L.
,
Laversanne
M.
,
Soerjomataram
I.
,
Jemal
A.
et al.
(
2021
)
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J. Clin.
71
,
209
249
[PubMed]
2.
Riis
M.
(
2020
)
Modern surgical treatment of breast cancer
.
Ann. Med. Surg.
56
,
95
107
3.
Yang
T.J.
and
Ho
A.Y.
(
2013
)
Radiation therapy in the management of breast cancer
.
Surg. Clin.
93
,
455
471
4.
Taghian
A.G.
and
Powell
S.N.
(
1999
)
The role of radiation therapy for primary breast cancer
.
Surg. Clin. North Am.
79
,
1091
1115
[PubMed]
5.
Moo
T.-A.
,
Sanford
R.
,
Dang
C.
and
Morrow
M.
(
2018
)
Overview of breast cancer therapy
.
PET Clin.
13
,
339
354
[PubMed]
6.
Hassan
M.
,
Ansari
J.
,
Spooner
D.
and
Hussain
S.
(
2010
)
Chemotherapy for breast cancer
.
Oncol. Rep.
24
,
1121
1131
[PubMed]
7.
Banerjee
D.
and
Sengupta
S.
(
2011
)
Nanoparticles in cancer chemotherapy
.
Prog. Mol. Biol. Transl. Sci.
104
,
489
507
[PubMed]
8.
Tinkle
S.
,
McNeil
S.E.
,
Mühlebach
S.
,
Bawa
R.
,
Borchard
G.
,
Barenholz
Y.
et al.
(
2014
)
Nanomedicines: addressing the scientific and regulatory gap
.
Ann. N. Y. Acad. Sci.
1313
,
35
56
[PubMed]
9.
Bayda
S.
,
Adeel
M.
,
Tuccinardi
T.
,
Cordani
M.
and
Rizzolio
F.
(
2019
)
The history of nanoscience and nanotechnology: from chemical–physical applications to nanomedicine
.
Molecules
25
,
112
[PubMed]
10.
Blanco
E.
,
Shen
H.
and
Ferrari
M.
(
2015
)
Principles of nanoparticle design for overcoming biological barriers to drug delivery
.
Nat. Biotechnol.
33
,
941
951
[PubMed]
11.
Davis
M.E.
,
Chen
Z.
and
Shin
D.M.
(
2008
)
Nanoparticle therapeutics: an emerging treatment modality for cancer
.
Nat. Rev. Drug Discovery
7
,
771
782
12.
Aslan
B.
,
Ozpolat
B.
,
Sood
A.K.
and
Lopez-Berestein
G.
(
2013
)
Nanotechnology in cancer therapy
.
J. Drug Target.
21
,
904
913
[PubMed]
13.
Sengupta
S.
(
2017
)
Cancer nanomedicine: lessons for immuno-oncology
.
Trends Cancer
3
,
551
560
[PubMed]
14.
Bartlett
D.W.
and
Davis
M.E.
(
2007
)
Physicochemical and biological characterization of targeted, nucleic acid-containing nanoparticles
.
Bioconjug. Chem.
18
,
456
468
[PubMed]
15.
Song
E.
,
Zhu
P.
,
Lee
S.-K.
,
Chowdhury
D.
,
Kussman
S.
,
Dykxhoorn
D.M.
et al.
(
2005
)
Antibody mediated in vivo delivery of small interfering RNAs via cell-surface receptors
.
Nat. Biotechnol.
23
,
709
717
[PubMed]
16.
Ediriwickrema
A.
and
Saltzman
W.M.
(
2015
)
Nanotherapy for cancer: targeting and multifunctionality in the future of cancer therapies
.
ACS Biomaterials Sci. Eng.
1
,
64
78
17.
Derakhshandeh
K.
and
Azandaryani
A.H.
(
2016
)
Active-targeted nanotherapy as smart cancer treatment
.
Smart Drug Deliv. Syst.
91
116
18.
Lee
J.H.
and
Nan
A.
(
2012
)
Combination drug delivery approaches in metastatic breast cancer
.
J. Drug Deliv.
2012
,
915375
19.
Wong
J.K.
,
Mohseni
R.
,
Hamidieh
A.A.
,
MacLaren
R.E.
,
Habib
N.
and
Seifalian
A.M.
(
2017
)
Will nanotechnology bring new hope for gene delivery?
Trends Biotechnol.
35
,
434
451
[PubMed]
20.
Nam
J.
,
Son
S.
,
Park
K.S.
,
Zou
W.
,
Shea
L.D.
and
Moon
J.J.
(
2019
)
Cancer nanomedicine for combination cancer immunotherapy
.
Nat. Rev. Mater.
4
,
398
414
21.
Baskar
R.
,
Lee
K.A.
,
Yeo
R.
and
Yeoh
K.-W.
(
2012
)
Cancer and radiation therapy: current advances and future directions
.
Int. J. Med. Sci.
9
,
193
22.
Yu
H.-j.
and
De Geest
B.G.
(
2020
)
Nanomedicine and cancer immunotherapy
,
41
,
Nature Publishing Group
23.
Van Hong Nguyen
B.-J.L.
(
2017
)
Protein corona: a new approach for nanomedicine design
.
Int. J. Nanomed.
12
,
3137
24.
Wilhelm
S.
,
Tavares
A.J.
,
Dai
Q.
,
Ohta
S.
,
Audet
J.
,
Dvorak
H.F.
et al.
(
2016
)
Analysis of nanoparticle delivery to tumours
.
Nat. Rev. Mater.
1
,
1
12
25.
Corbo
C.
,
Molinaro
R.
,
Parodi
A.
,
Toledano Furman
N.E.
,
Salvatore
F.
et al.
(
2016
)
The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery
.
Nanomedicine
11
,
81
100
[PubMed]
26.
Banerjee
S.S.
,
Aher
N.
,
Patil
R.
and
Khandare
J.
(
2012
)
Poly (ethylene glycol)-prodrug conjugates: concept, design, and applications
.
J. Drug Deliv.
2012
,
103973
27.
Suk
J.S.
,
Xu
Q.
,
Kim
N.
,
Hanes
J.
and
Ensign
L.M.
(
2016
)
PEGylation as a strategy for improving nanoparticle-based drug and gene delivery
.
Adv. Drug. Deliv. Rev.
99
,
28
51
[PubMed]
28.
Basu
S.
,
Chaudhuri
P.
and
Sengupta
S.
(
2009
)
Targeting oncogenic signaling pathways by exploiting nanotechnology
.
Cell Cycle
8
,
3480
3487
[PubMed]
29.
Attia
M.F.
,
Anton
N.
,
Wallyn
J.
,
Omran
Z.
and
Vandamme
T.F.
(
2019
)
An overview of active and passive targeting strategies to improve the nanocarriers efficiency to tumour sites
.
J. Pharm. Pharmacol.
71
,
1185
1198
[PubMed]
30.
Matsumura
Y.
and
Maeda
H.
(
1986
)
A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs
.
Cancer Res.
46
,
6387
6392
[PubMed]
31.
Sutton
D.
,
Nasongkla
N.
,
Blanco
E.
and
Gao
J.
(
2007
)
Functionalized micellar systems for cancer targeted drug delivery
.
Pharm. Res.
24
,
1029
1046
[PubMed]
32.
Sparreboom
A.
,
Scripture
C.D.
,
Trieu
V.
,
Williams
P.J.
,
De
T.
,
Yang
A.
et al.
(
2005
)
Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol)
.
Clin. Cancer Res.
11
,
4136
4143
[PubMed]
33.
Boddy
A.V.
,
Plummer
E.R.
,
Todd
R.
,
Sludden
J.
,
Griffin
M.
,
Robson
L.
et al.
(
2005
)
A phase I and pharmacokinetic study of paclitaxel poliglumex (XYOTAX), investigating both 3-weekly and 2-weekly schedules
.
Clin. Cancer Res.
11
,
7834
7840
[PubMed]
34.
Kraut
E.H.
,
Fishman
M.N.
,
Lorusso
P.M.
,
Gordon
M.S.
,
Rubin
E.H.
,
Haas
A.
et al.
(
2005
)
Final results of a phase I study of liposome encapsulated SN-38 (LE-SN38): safety, pharmacogenomics, pharmacokinetics, and tumor response
.
J. Clin. Oncol.
23
,
2017
2017
35.
Daud
A.
,
Garrett
C.
,
Simon
G.
,
Munster
P.
,
Sullivan
D.
,
Stromatt
S.
et al.
(
2006
)
Phase I trial of CT-2106 (polyglutamated camptothecin) administered weekly in patients (pts) with advanced solid tumors
.
J. Clin. Oncol.
24
,
2015
2015
36.
Yen
Y.
,
Synold
T.
,
Schluep
T.
,
Hwang
J.
,
Oliver
J.
and
Davis
M.
(
2007
)
First-in-human phase I trial of a cyclodextrin-containing polymer-camptothecin nanoparticle in patients with solid tumors
.
J. Clin. Oncol.
25
,
14078
14078
37.
Gao
F.
,
Zhang
Z.
,
Bu
H.
,
Huang
Y.
,
Gao
Z.
,
Shen
J.
et al.
(
2011
)
Nanoemulsion improves the oral absorption of candesartan cilexetil in rats: performance and mechanism
.
J. Control. Release
149
,
168
174
[PubMed]
38.
Ganassin
R.
,
Merker
C.
,
Rodrigues
M.C.
,
Guimarães
N.F.
,
Sodré
C.S.C.
,
Ferreira
Q.d.S.
et al.
(
2018
)
Nanocapsules for the co-delivery of selol and doxorubicin to breast adenocarcinoma 4T1 cells in vitro
.
Artificial Cells Nanomed. Biotechnol.
46
,
2002
2012
39.
Sun
W.
,
Jiang
T.
,
Lu
Y.
,
Reiff
M.
,
Mo
R.
and
Gu
Z.
(
2014
)
Cocoon-like self-degradable DNA nanoclew for anticancer drug delivery
.
J. Am. Chem. Soc.
136
,
14722
14725
[PubMed]
40.
Dong
X.
,
Sun
Z.
,
Wang
X.
and
Leng
X.
(
2017
)
An innovative MWCNTs/DOX/TC nanosystem for chemo-photothermal combination therapy of cancer
.
Nanomed. Nanotechnol. Biol. Med.
13
,
2271
2280
41.
Muhamad
N.
,
Plengsuriyakarn
T.
and
Na-Bangchang
K.
(
2018
)
Application of active targeting nanoparticle delivery system for chemotherapeutic drugs and traditional/herbal medicines in cancer therapy: a systematic review
.
Int. J. Nanomed.
13
,
3921
42.
Gray
B.P.
and
Brown
K.C.
(
2014
)
Combinatorial peptide libraries: mining for cell-binding peptides
.
Chem. Rev.
114
,
1020
1081
[PubMed]
43.
Bross
P.F.
,
Beitz
J.
,
Chen
G.
,
Chen
X.H.
,
Duffy
E.
,
Kieffer
L.
et al.
(
2001
)
Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia
.
Clin. Cancer Res.
7
,
1490
1496
[PubMed]
44.
Kawakami
K.
,
Nakajima
O.
,
Morishita
R.
and
Nagai
R.
(
2006
)
Targeted anticancer immunotoxins and cytotoxic agents with direct killing moieties
.
Sci. World J.
6
,
781
790
45.
Allen
T.M.
(
2002
)
Ligand-targeted therapeutics in anticancer therapy
.
Nat. Rev. Cancer
2
,
750
763
[PubMed]
46.
Duncan
R.
(
2006
)
Polymer conjugates as anticancer nanomedicines
.
Nat. Rev. Cancer
6
,
688
701
[PubMed]
47.
Matsumura
Y.
,
Gotoh
M.
,
Muro
K.
,
Yamada
Y.
,
Shirao
K.
,
Shimada
Y.
et al.
(
2004
)
Phase I and pharmacokinetic study of MCC-465, a doxorubicin (DXR) encapsulated in PEG immunoliposome, in patients with metastatic stomach cancer
.
Ann. Oncol.
15
,
517
525
[PubMed]
48.
Phan
A.
,
Takimoto
C.
,
Adinin
R.
,
Wood
L.
,
Xiong
H.
,
Matsuno
K.
et al.
(
2007
)
Open label phase I study of MBP-426, a novel formulation of oxaliplatin, in patients with advanced or metastatic solid tumors
.
AACR
6
,
C115
49.
Tang
W.
,
Lalezari
J.
,
June
C.
,
Tebas
P.
,
Lee
G.
,
Giedlin
M.
et al.
(
2012
)
51. Results of a Phase I Trial of SGT-53: A Systemically Administered, Tumor-Targeting Immunoliposome Nanocomplex Incorporating a Plasmid Encoding wtp53
.
Mol. Ther.
20
,
1
[PubMed]
50.
Heidel
J.D.
,
Yu
Z.
,
Liu
J.Y.-C.
,
Rele
S.M.
,
Liang
Y.
,
Zeidan
R.K.
et al.
(
2007
)
Administration in non-human primates of escalating intravenous doses of targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA
.
Proc. Natl. Acad. Sci.
104
,
5715
5721
51.
Autio
K.A.
,
Dreicer
R.
,
Anderson
J.
,
Garcia
J.A.
,
Alva
A.
,
Hart
L.L.
et al.
(
2018
)
Safety and efficacy of BIND-014, a docetaxel nanoparticle targeting prostate-specific membrane antigen for patients with metastatic castration-resistant prostate cancer: a phase 2 clinical trial
.
JAMA Oncol.
4
,
1344
1351
[PubMed]
52.
Cultrara
C.N.
,
Shah
S.
,
Antuono
G.
,
Heller
C.J.
,
Ramos
J.A.
,
Samuni
U.
et al.
(
2019
)
Size matters: arginine-derived peptides targeting the PSMA receptor can efficiently complex but not transfect siRNA
.
Mol. Therapy-Nucleic Acids
18
,
863
870
53.
Lee
J.H.
,
Engler
J.A.
,
Collawn
J.F.
and
Moore
B.A.
(
2001
)
Receptor mediated uptake of peptides that bind the human transferrin receptor
.
Eur. J. Biochem.
268
,
2004
2012
[PubMed]
54.
Worm
D.J.
,
Els‐Heindl
S.
and
Beck‐Sickinger
A.G.
(
2020
)
Targeting of peptide‐binding receptors on cancer cells with peptide‐drug conjugates
.
Pept. Sci.
112
,
e24171
55.
Sun
L.-C.
,
;
H.
and
Coy
D.
(
2011
)
Somatostatin receptor-targeted anti-cancer therapy
.
Curr. Drug Deliv.
8
,
2
10
56.
Sancho
V.
,
Di Florio
A.
,
;
W.
,
Moody
T.
,
;
T.
and
Jensen
R.
(
2011
)
Bombesin receptor-mediated imaging and cytotoxicity: review and current status
.
Curr. Drug Deliv.
8
,
79
134
57.
Kokko
K.P.
,
Hadden
M.K.
,
Orwig
K.S.
,
Mazella
J.
and
Dix
T.A.
(
2003
)
In vitro analysis of stable, receptor-selective neurotensin [8− 13] analogues
.
J. Med. Chem.
46
,
4141
4148
[PubMed]
58.
Froidevaux
S.
,
Calame-Christe
M.
,
Tanner
H.
and
Eberle
A.N.
(
2005
)
Melanoma targeting with DOTA-α-melanocyte-stimulating hormone analogs: structural parameters affecting tumor uptake and kidney uptake
.
J. Nucl. Med.
46
,
887
895
[PubMed]
59.
Courcier
J.
,
de la Taille
A.
,
Nourieh
M.
,
Leguerney
I.
,
Lassau
N.
and
Ingels
A.
(
2020
)
Carbonic Anhydrase IX in renal cell carcinoma, implications for disease management
.
Int. J. Mol. Sci.
21
,
7146
60.
Friedman
A.D.
,
Claypool
S.E.
and
Liu
R.
(
2013
)
The smart targeting of nanoparticles
.
Curr. Pharm. Des.
19
,
6315
6329
[PubMed]
61.
Werengowska-Ciećwierz
K.
,
Wiśniewski
M.
,
Terzyk
A.P.
and
Furmaniak
S.
(
2015
)
The chemistry of bioconjugation in nanoparticles-based drug delivery system
.
Adv. Condens. Matter Phys.
2015
,
198175
62.
Toy
R.
,
Peiris
P.M.
,
Ghaghada
K.B.
and
Karathanasis
E.
(
2014
)
Shaping cancer nanomedicine: the effect of particle shape on the in vivo journey of nanoparticles
.
Nanomedicine
9
,
121
134
[PubMed]
63.
Albanese
A.
,
Tang
P.S.
and
Chan
W.C.
(
2012
)
The effect of nanoparticle size, shape, and surface chemistry on biological systems
.
Annu. Rev. Biomed. Eng.
14
,
1
16
[PubMed]
64.
Stokes
L.D.
Jr
(
2021
)
Immunomodulatory biomaterials for cancer immunotherapy
.
Honors Theses
1767
,
https://egrove.olemiss.edu/hon_thesis/1767
65.
Sykes
E.A.
,
Dai
Q.
,
Sarsons
C.D.
,
Chen
J.
,
Rocheleau
J.V.
,
Hwang
D.M.
et al.
(
2016
)
Tailoring nanoparticle designs to target cancer based on tumor pathophysiology
.
Proc. Natl. Acad. Sci.
113
,
E1142
E1151
66.
Petros
R.A.
and
DeSimone
J.M.
(
2010
)
Strategies in the design of nanoparticles for therapeutic applications
.
Nat. Rev. Drug Discov.
9
,
615
627
67.
Doshi
N.
,
Zahr
A.S.
,
Bhaskar
S.
,
Lahann
J.
and
Mitragotri
S.
(
2009
)
Red blood cell-mimicking synthetic biomaterial particles
.
Proc. Natl. Acad. Sci.
106
,
21495
21499
68.
Malhotra
S.
,
Dumoga
S.
,
Sirohi
P.
and
Singh
N.
(
2019
)
Red blood cells-derived vesicles for delivery of lipophilic drug camptothecin
.
ACS Appl. Mater. Interfaces
11
,
22141
22151
69.
Wang
D.
,
Dong
H.
,
Li
M.
,
Cao
Y.
,
Yang
F.
,
Zhang
K.
et al.
(
2018
)
Erythrocyte–cancer hybrid membrane camouflaged hollow copper sulfide nanoparticles for prolonged circulation life and homotypic-targeting photothermal/chemotherapy of melanoma
.
ACS Nano
12
,
5241
5252
[PubMed]
70.
Chou
L.Y.
,
Ming
K.
and
Chan
W.C.
(
2011
)
Strategies for the intracellular delivery of nanoparticles
.
Chem. Soc. Rev.
40
,
233
245
[PubMed]
71.
Jiang
W.
,
Kim
B.Y.
,
Rutka
J.T.
and
Chan
W.C.
(
2008
)
Nanoparticle-mediated cellular response is size-dependent
.
Nat. Nanotechnol.
3
,
145
150
[PubMed]
72.
Huang
X.
and
Brazel
C.S.
(
2001
)
On the importance and mechanisms of burst release in matrix-controlled drug delivery systems
.
J. Control. Release
73
,
121
136
[PubMed]
73.
Simonazzi
A.
,
Cid
A.G.
,
Villegas
M.
,
Romero
A.I.
,
Palma
S.D.
and
Bermúdez
J.M.
(
2018
)
Nanotechnology applications in drug controlled release
.
Drug targeting and stimuli sensitive drug delivery systems
, pp.
81
116
,
Elsevier
74.
Kamaly
N.
,
Yameen
B.
,
Wu
J.
and
Farokhzad
O.C.
(
2016
)
Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release
.
Chem. Rev.
116
,
2602
2663
[PubMed]
75.
Lu
B.
,
Lv
X.
and
Le
Y.
(
2019
)
Chitosan-modified PLGA nanoparticles for control-released drug delivery
.
Polymers
11
,
304
76.
Jang
H.L.
and
Sengupta
S.
(
2019
)
Transcellular transfer of nanomedicine
.
Nat. Nanotechnol.
14
,
731
732
[PubMed]
77.
Paraskar
A.
,
Soni
S.
,
Basu
S.
,
Amarasiriwardena
C.J.
,
Lupoli
N.
,
Srivats
S.
et al.
(
2011
)
Rationally engineered polymeric cisplatin nanoparticles for improved antitumor efficacy
.
Nanotechnology
22
,
265101
[PubMed]
78.
Paraskar
A.S.
,
Soni
S.
,
Chin
K.T.
,
Chaudhuri
P.
,
Muto
K.W.
,
Berkowitz
J.
et al.
(
2010
)
Harnessing structure-activity relationship to engineer a cisplatin nanoparticle for enhanced antitumor efficacy
.
Proc. Natl. Acad. Sci.
107
,
12435
12440
79.
Sengupta
P.
,
Basu
S.
,
Soni
S.
,
Pandey
A.
,
Roy
B.
,
Oh
M.S.
et al.
(
2012
)
Cholesterol-tethered platinum II-based supramolecular nanoparticle increases antitumor efficacy and reduces nephrotoxicity
.
Proc. Natl. Acad. Sci.
109
,
11294
11299
80.
Paraskar
A.
,
Soni
S.
,
Roy
B.
,
Papa
A.-L.
and
Sengupta
S.
(
2012
)
Rationally designed oxaliplatin-nanoparticle for enhanced antitumor efficacy
.
Nanotechnology
23
,
075103
[PubMed]
81.
Kulkarni
A.
,
Pandey
P.
,
Rao
P.
,
Mahmoud
A.
,
Goldman
A.
,
Sabbisetti
V.
et al.
(
2016
)
Algorithm for designing nanoscale supramolecular therapeutics with increased anticancer efficacy
.
ACS Nano
10
,
8154
8168
[PubMed]
82.
Huang
L.
and
Liu
Y.
(
2011
)
In vivo delivery of RNAi with lipid-based nanoparticles
.
Annu. Rev. Biomed. Eng.
13
,
507
530
[PubMed]
83.
Mishra
J.
and
Panda
J.J.
(
2019
)
Short peptide-based smart targeted cancer nanotherapeutics: a glimmer of hope
.
Future Sci.
84.
Tai
W.
and
Gao
X.
(
2017
)
Functional peptides for siRNA delivery
.
Adv. Drug. Deliv. Rev.
110
,
157
168
[PubMed]
85.
Dutta
C.
,
Chakraborty
K.
and
Sinha Roy
R.
(
2015
)
Engineered nanostructured facial lipopeptide as highly efficient molecular transporter
.
ACS Appl. Mater. Interfaces
7
,
18397
18405
86.
Biswas
A.
,
Chakraborty
K.
,
Dutta
C.
,
Mukherjee
S.
,
Gayen
P.
,
Jan
S.
et al.
(
2019
)
Engineered histidine-enriched facial Lipopeptides for enhanced intracellular delivery of functional siRNA to triple negative breast Cancer cells
.
ACS Appl. Mater. Interfaces
11
,
4719
4736
87.
Kim
B.Y.
,
Rutka
J.T.
and
Chan
W.C.
(
2010
)
Nanomedicine
.
N. Engl. J. Med.
363
,
2434
2443
[PubMed]
88.
Kulkarni
A.A.
,
Vijaykumar
V.E.
,
Natarajan
S.K.
,
Sengupta
S.
and
Sabbisetti
V.S.
(
2016
)
Sustained inhibition of cMET-VEGFR2 signaling using liposome-mediated delivery increases efficacy and reduces toxicity in kidney cancer
.
Nanomed. Nanotechnol. Biol. Med.
12
,
1853
1861
89.
Basu
S.
,
Harfouche
R.
,
Soni
S.
,
Chimote
G.
,
Mashelkar
R.A.
and
Sengupta
S.
(
2009
)
Nanoparticle-mediated targeting of MAPK signaling predisposes tumor to chemotherapy
.
Proc. Natl. Acad. Sci.
106
,
7957
7961
90.
Harfouche
R.
,
Basu
S.
,
Soni
S.
,
Hentschel
D.M.
,
Mashelkar
R.A.
and
Sengupta
S.
(
2009
)
Nanoparticle-mediated targeting of phosphatidylinositol-3-kinase signaling inhibits angiogenesis
.
Angiogenesis
12
,
325
[PubMed]
91.
Kulkarni
A.A.
,
Roy
B.
,
Rao
P.S.
,
Wyant
G.A.
,
Mahmoud
A.
,
Ramachandran
M.
et al.
(
2013
)
Supramolecular nanoparticles that target phosphoinositide-3-kinase overcome insulin resistance and exert pronounced antitumor efficacy
.
Cancer Res.
73
,
6987
6997
[PubMed]
92.
Pandey
A.
,
Kulkarni
A.
,
Roy
B.
,
Goldman
A.
,
Sarangi
S.
,
Sengupta
P.
et al.
(
2014
)
Sequential application of a cytotoxic nanoparticle and a PI3K inhibitor enhances antitumor efficacy
.
Cancer Res.
74
,
675
685
[PubMed]
93.
Hoffmann
F.
,
Sass
G.
,
Zillies
J.
,
Zahler
S.
,
Tiegs
G.
,
Hartkorn
A.
et al.
(
2009
)
A novel technique for selective NF-κB inhibition in Kupffer cells: contrary effects in fulminant hepatitis and ischaemia–reperfusion
.
Gut
58
,
1670
1678
[PubMed]
94.
Hattori
Y.
,
Sakaguchi
M.
and
Maitani
Y.
(
2006
)
Folate-linked lipid-based nanoparticles deliver a NFκB decoy into activated murine macrophage-like RAW264. 7 cells
.
Biol. Pharm. Bull.
29
,
1516
1520
[PubMed]
95.
Xu
M.-X.
,
Ge
C.-X.
,
Li
Q.
,
Lou
D.-S.
,
Hu
L.-F.
,
Sun
Y.
et al.
(
2020
)
Fisetin nanoparticles protect against PM2. 5 exposure-induced neuroinflammation by down-regulation of astrocytes activation related NF-κB signaling pathway
.
J. Funct. Foods
65
,
103716
96.
Gai
W.
,
Hao
X.
,
Zhao
J.
,
Wang
L.
,
Liu
J.
,
Jiang
H.
et al.
(
2020
)
Delivery of benzoylaconitine using biodegradable nanoparticles to suppress inflammation via regulating NF-κB signaling
.
Colloids Surf. B
191
,
110980
97.
Chen
Y.-P.
,
Chen
C.-T.
,
Liu
T.-P.
,
Chien
F.-C.
,
Wu
S.-H.
,
Chen
P.
et al.
(
2020
)
Catcher in the rel: nanoparticles-antibody conjugate as NF-κB nuclear translocation blocker
.
Biomaterials
246
,
119997
[PubMed]
98.
Abdel-Hakeem
E.A.
,
Abdel-Hamid
H.A.
and
Abdel Hafez
S.M.N.
(
2020
)
The possible protective effect of Nano-Selenium on the endocrine and exocrine pancreatic functions in a rat model of acute pancreatitis
.
J. Trace Elem. Med. Biol.
60
,
126480
[PubMed]
99.
Xu
X.
,
Rui
S.
,
Chen
C.
,
Zhang
G.
,
Li
Z.
,
Wang
J.
et al.
(
2020
)
Protective effects of astragalus polysaccharide nanoparticles on septic cardiac dysfunction through inhibition of TLR4/NF-κB signaling pathway
.
Int. J. Biol. Macromol.
153
,
977
985
[PubMed]
100.
Sahab-Negah
S.
,
Ariakia
F.
,
Jalili-Nik
M.
,
Afshari
A.R.
,
Salehi
S.
,
Samini
F.
et al.
(
2020
)
Curcumin loaded in niosomal nanoparticles improved the anti-tumor effects of free curcumin on glioblastoma stem-like cells: an in vitro study
.
Mol. Neurobiol.
57
,
3391
3411
[PubMed]
101.
Ramirez
R.
,
Matrana
M.
,
Satti
S.
,
Griffin
R.
,
Voros
B.
,
Mohammed
A.
et al.
(
2019
)
EP1.12-18 NET-001: A Phase II Study of ABI-009 in Metastatic Neuroendocrine Tumors of the Lung or Gastroenteropancreatic System
.
J. Thoracic Oncol.
14
,
102.
Mamaeva
V.
,
Rosenholm
J.M.
,
Bate-Eya
L.T.
,
Bergman
L.
,
Peuhu
E.
,
Duchanoy
A.
et al.
(
2011
)
Mesoporous silica nanoparticles as drug delivery systems for targeted inhibition of Notch signaling in cancer
.
Mol. Ther.
19
,
1538
1546
[PubMed]
103.
Tsai
Y.S.
,
Chen
Y.H.
,
Cheng
P.C.
,
Tsai
H.T.
,
Shiau
A.L.
,
Tzai
T.S.
et al.
(
2013
)
TGF‐β1 Conjugated to gold nanoparticles results in protein conformational changes and attenuates the biological function
.
Small
9
,
2119
2128
[PubMed]
104.
Verma
R.K.
,
Yu
W.
,
Singh
S.P.
,
Shankar
S.
and
Srivastava
R.K.
(
2015
)
Anthothecol-encapsulated PLGA nanoparticles inhibit pancreatic cancer stem cell growth by modulating sonic hedgehog pathway
.
Nanomed. Nanotechnol. Biol. Med.
11
,
2061
2070
105.
Crombez
L.
,
Morris
M.C.
,
Dufort
S.
,
Aldrian-Herrada
G.
,
Nguyen
Q.
,
Mc Master
G.
et al.
(
2009
)
Targeting cyclin B1 through peptide-based delivery of siRNA prevents tumour growth
.
Nucleic Acids Res.
37
,
4559
4569
[PubMed]
106.
Kanazawa
T.
,
Morisaki
K.
,
Suzuki
S.
and
Takashima
Y.
(
2014
)
Prolongation of life in rats with malignant glioma by intranasal siRNA/drug codelivery to the brain with cell-penetrating peptide-modified micelles
.
Mol. Pharm.
11
,
1471
1478
[PubMed]
107.
Lee
S.H.
,
Kim
S.H.
and
Park
T.G.
(
2007
)
Intracellular siRNA delivery system using polyelectrolyte complex micelles prepared from VEGF siRNA-PEG conjugate and cationic fusogenic peptide
.
Biochem. Biophys. Res. Commun.
357
,
511
516
[PubMed]
108.
Gayen
P.
,
Jan
S.
,
Chowdhury
N.
,
Ghosh
S.
,
Hembram
M.
,
Bagchi
A.
et al.
(
2021
)
Engineered bio‐inspired multifunctional peptide‐and protein‐based therapeutic biomolecules for better wound care
.
Chemistry–An Asian J.
16
,
4018
4036
109.
Ghosh
S.
,
Gayen
P.
,
Jan
S.
,
Kishore
A.V.
,
Kumar
V.
,
Mallick
A.M.
et al.
(
2020
)
Bioinspired non-immunogenic multifunctional sealant for efficient blood clotting and suture-free wound closure
.
ACS Biomaterials Sci. Eng.
6
,
6378
6393
110.
Ghosh
S.
,
Mukherjee
S.
,
Dutta
C.
,
Chakraborty
K.
,
Gayen
P.
,
Jan
S.
et al.
(
2017
)
Engineered isopeptide bond stabilized fibrin inspired nanoscale peptide based sealants for efficient blood clotting
.
Sci. Rep.
7
,
1
13
[PubMed]
111.
Ghosh
S.
,
Tripathi
A.
,
Gayen
P.
and
Roy
R.S.
(
2020
)
Peptide-based topical agents and intravenous hemostat for rapid hemostasis
.
RSC Medicinal Chem.
11
,
1100
1111
112.
Nishida
N.
,
Yano
H.
,
Nishida
T.
,
Kamura
T.
and
Kojiro
M.
(
2006
)
Angiogenesis in cancer
.
Vascular Health Risk Manage.
2
,
213
113.
Banerjee
D.
,
Harfouche
R.
and
Sengupta
S.
(
2011
)
Nanotechnology-mediated targeting of tumor angiogenesis
.
Vascular Cell
3
,
1
13
[PubMed]
114.
Kerbel
R.S.
and
Kamen
B.A.
(
2004
)
The anti-angiogenic basis of metronomic chemotherapy
.
Nat. Rev. Cancer
4
,
423
436
[PubMed]
115.
Sengupta
S.
,
Eavarone
D.
,
Capila
I.
,
Zhao
G.
,
Watson
N.
,
Kiziltepe
T.
et al.
(
2005
)
Temporal targeting of tumour cells and neovasculature with a nanoscale delivery system
.
Nature
436
,
568
572
[PubMed]
116.
Chaudhuri
P.
,
Harfouche
R.
,
Soni
S.
,
Hentschel
D.M.
and
Sengupta
S.
(
2010
)
Shape effect of carbon nanovectors on angiogenesis
.
Acs Nano
4
,
574
582
[PubMed]
117.
Satchi-Fainaro
R.
,
Puder
M.
,
Davies
J.W.
,
Tran
H.T.
,
Sampson
D.A.
,
Greene
A.K.
et al.
(
2004
)
Targeting angiogenesis with a conjugate of HPMA copolymer and TNP-470
.
Nat. Med.
10
,
255
261
[PubMed]
118.
Miller
K.
,
Erez
R.
,
Segal
E.
,
Shabat
D.
and
Satchi‐Fainaro
R.
(
2009
)
Targeting bone metastases with a bispecific anticancer and antiangiogenic polymer–alendronate–taxane conjugate
.
Angew. Chem.
121
,
2993
2998
119.
Mitra
A.
,
Mulholland
J.
,
Nan
A.
,
McNeill
E.
,
Ghandehari
H.
and
Line
B.R.
(
2005
)
Targeting tumor angiogenic vasculature using polymer–RGD conjugates
.
J. Control. Release
102
,
191
201
[PubMed]
120.
Kim
W.J.
,
Yockman
J.W.
,
Jeong
J.H.
,
Christensen
L.V.
,
Lee
M.
,
Kim
Y.-H.
et al.
(
2006
)
Anti-angiogenic inhibition of tumor growth by systemic delivery of PEI-g-PEG-RGD/pCMV-sFlt-1 complexes in tumor-bearing mice
.
J. Control. Release
114
,
381
388
[PubMed]
121.
Schiffelers
R.M.
,
Ansari
A.
,
Xu
J.
,
Zhou
Q.
,
Tang
Q.
,
Storm
G.
et al.
(
2004
)
Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle
.
Nucleic Acids Res.
32
,
e149
e149
[PubMed]
122.
Pillé
J.-Y.
,
Li
H.
,
Blot
E.
,
Bertrand
J.-R.
,
Pritchard
L.-L.
,
Opolon
P.
et al.
(
2006
)
Intravenous delivery of anti-RhoA small interfering RNA loaded in nanoparticles of chitosan in mice: safety and efficacy in xenografted aggressive breast cancer
.
Hum. Gene Ther.
17
,
1019
1026
[PubMed]
123.
Backer
M.V.
,
Gaynutdinov
T.I.
,
Patel
V.
,
Bandyopadhyaya
A.K.
,
Thirumamagal
B.
,
Tjarks
W.
et al.
(
2005
)
Vascular endothelial growth factor selectively targets boronated dendrimers to tumor vasculature
.
Mol. Cancer Ther.
4
,
1423
1429
[PubMed]
124.
Benny
O.
,
Fainaru
O.
,
Adini
A.
,
Cassiola
F.
,
Bazinet
L.
,
Adini
I.
et al.
(
2008
)
An orally delivered small-molecule formulation with antiangiogenic and anticancer activity
.
Nat. Biotechnol.
26
,
799
807
[PubMed]
125.
Nasongkla
N.
,
Shuai
X.
,
Ai
H.
,
Weinberg
B.D.
,
Pink
J.
,
Boothman
D.A.
et al.
(
2004
)
cRGD‐functionalized polymer micelles for targeted doxorubicin delivery
.
Angew. Chem.
116
,
6483
6487
126.
Maeda
N.
,
Takeuchi
Y.
,
Takada
M.
,
Sadzuka
Y.
,
Namba
Y.
and
Oku
N.
(
2004
)
Anti-neovascular therapy by use of tumor neovasculature-targeted long-circulating liposome
.
J. Control. Release
100
,
41
52
[PubMed]
127.
Villares
G.J.
,
Zigler
M.
,
Wang
H.
,
Melnikova
V.O.
,
Wu
H.
,
Friedman
R.
et al.
(
2008
)
Targeting melanoma growth and metastasis with systemic delivery of liposome-incorporated protease-activated receptor-1 small interfering RNA
.
Cancer Res.
68
,
9078
9086
[PubMed]
128.
Chen
J.
,
Wu
H.
,
Han
D.
and
Xie
C.
(
2006
)
Using anti-VEGF McAb and magnetic nanoparticles as double-targeting vector for the radioimmunotherapy of liver cancer
.
Cancer Lett.
231
,
169
175
[PubMed]
129.
Maeng
J.H.
,
Lee
D.-H.
,
Jung
K.H.
,
Bae
Y.-H.
,
Park
I.-S.
,
Jeong
S.
et al.
(
2010
)
Multifunctional doxorubicin loaded superparamagnetic iron oxide nanoparticles for chemotherapy and magnetic resonance imaging in liver cancer
.
Biomaterials
31
,
4995
5006
[PubMed]
130.
Cui
T.
,
Liang
J.-J.
,
Chen
H.
,
Geng
D.-D.
,
Jiao
L.
,
Yang
J.-Y.
et al.
(
2017
)
Performance of doxorubicin-conjugated gold nanoparticles: regulation of drug location
.
ACS Appl. Mater. Interfaces
9
,
8569
8580
131.
Mirza
Z.
and
Karim
S.
(
2021
)
Nanoparticles-based drug delivery and gene therapy for breast cancer: Recent advancements and future challenges
.
Seminars in cancer biology
, pp.
226
237
,
Elsevier
132.
Yin
H.
,
Kanasty
R.L.
,
Eltoukhy
A.A.
,
Vegas
A.J.
,
Dorkin
J.R.
and
Anderson
D.G.
(
2014
)
Non-viral vectors for gene-based therapy
.
Nat. Rev. Genet.
15
,
541
555
[PubMed]
133.
Wang
K.
,
Kievit
F.M.
and
Zhang
M.
(
2016
)
Nanoparticles for cancer gene therapy: Recent advances, challenges, and strategies
.
Pharmacol. Res.
114
,
56
66
[PubMed]
134.
Lonez
C.
,
Vandenbranden
M.
and
Ruysschaert
J.-M.
(
2008
)
Cationic liposomal lipids: from gene carriers to cell signaling
.
Prog. Lipid Res.
47
,
340
347
[PubMed]
135.
Kanasty
R.
,
Dorkin
J.R.
,
Vegas
A.
and
Anderson
D.
(
2013
)
Delivery materials for siRNA therapeutics
.
Nat. Mater.
12
,
967
977
[PubMed]
136.
Hou
X.
,
Zaks
T.
,
Langer
R.
and
Dong
Y.
(
2021
)
Lipid nanoparticles for mRNA delivery
.
Nat. Rev. Mater.
6
,
1078
1094
[PubMed]
137.
Wang
X.
,
Niu
D.
,
Hu
C.
and
Li
P.
(
2015
)
Polyethyleneimine-based nanocarriers for gene delivery
.
Curr. Pharm. Des.
21
,
6140
6156
[PubMed]
138.
Ekladious
I.
,
Colson
Y.L.
and
Grinstaff
M.W.
(
2019
)
Polymer–drug conjugate therapeutics: advances, insights and prospects
.
Nat. Rev. Drug Discovery
18
,
273
294
139.
Appelbaum
J.S.
,
LaRochelle
J.R.
,
Smith
B.A.
,
Balkin
D.M.
,
Holub
J.M.
and
Schepartz
A.
(
2012
)
Arginine topology controls escape of minimally cationic proteins from early endosomes to the cytoplasm
.
Chem. Biol.
19
,
819
830
[PubMed]
140.
Mallick
A.M.
,
Chakraborty
K.
,
Biswas
A.
,
Jan
S.
,
Dutta
C.
,
Dey
S.
et al.
(
2019
)
Emerging peptide-based technologies in cancer therapy
.
Unravelling Cancer Signaling Pathways: A Multidisciplinary Approach
, pp.
13
49
,
Springer
141.
Bulut
S.
,
Erkal
T.S.
,
Toksoz
S.
,
Tekinay
A.B.
,
Tekinay
T.
and
Guler
M.O.
(
2011
)
Slow release and delivery of antisense oligonucleotide drug by self-assembled peptide amphiphile nanofibers
.
Biomacromolecules
12
,
3007
3014
[PubMed]
142.
Mazza
M.
,
Hadjidemetriou
M.
,
de Lazaro
I.
,
Bussy
C.
and
Kostarelos
K.
(
2015
)
Peptide nanofiber complexes with siRNA for deep brain gene silencing by stereotactic neurosurgery
.
ACS Nano
9
,
1137
1149
[PubMed]
143.
Deshayes
S.
,
Morris
M.
,
Divita
G.
and
Heitz
F.
(
2005
)
Cell-penetrating peptides: tools for intracellular delivery of therapeutics
.
Cell. Mol. Life Sci. CMLS
62
,
1839
1849
144.
Deshayes
S.
,
Gerbal-Chaloin
S.
,
Morris
M.C.
,
Aldrian-Herrada
G.
,
Charnet
P.
,
Divita
G.
et al.
(
2004
)
On the mechanism of non-endosomial peptide-mediated cellular delivery of nucleic acids
.
Biochimica et Biophysica Acta
1667
,
141
147
145.
Tarvirdipour
S.
,
Huang
X.
,
Mihali
V.
,
Schoenenberger
C.-A.
and
Palivan
C.G.
(
2020
)
Peptide-based nanoassemblies in gene therapy and diagnosis: paving the way for clinical application
.
Molecules
25
,
3482
146.
Dizaj
S.M.
,
Jafari
S.
and
Khosroushahi
A.Y.
(
2014
)
A sight on the current nanoparticle-based gene delivery vectors
.
Nanoscale Res. Lett.
9
,
1
9
[PubMed]
147.
Pantarotto
D.
,
Briand
J.-P.
,
Prato
M.
and
Bianco
A.
(
2004
)
Translocation of bioactive peptides across cell membranes by carbon nanotubes
.
Chem. Commun.
16
17
148.
Isobe
H.
,
Nakanishi
W.
,
Tomita
N.
,
Jinno
S.
,
Okayama
H.
and
Nakamura
E.
(
2006
)
Gene delivery by aminofullerenes: structural requirements for efficient transfection
.
Chemistry–An Asian J.
1
,
167
175
149.
Chen
Y.
,
Chen
H.
and
Shi
J.
(
2014
)
Inorganic nanoparticle-based drug codelivery nanosystems to overcome the multidrug resistance of cancer cells
.
Mol. Pharm.
11
,
2495
2510
[PubMed]
150.
Hattab
D.
,
Gazzali
A.M.
and
Bakhtiar
A.
(
2021
)
Clinical advances of siRNA-based nanotherapeutics for cancer treatment
.
Pharmaceutics
13
,
1009
[PubMed]
151.
Hattab
D.
and
Bakhtiar
A.
(
2020
)
Bioengineered siRNA-based nanoplatforms targeting molecular signaling pathways for the treatment of triple negative breast cancer: preclinical and clinical advancements
.
Pharmaceutics
12
,
929
152.
Salvioni
L.
,
Zuppone
S.
,
Andreata
F.
,
Monieri
M.
,
Mazzucchelli
S.
,
Di Carlo
C.
et al.
(
2020
)
Nanoparticle‐mediated suicide gene therapy for triple negative breast cancer treatment
.
Adv. Therapeut.
3
,
2000007
153.
Choi
J.
,
Rui
Y.
,
Kim
J.
,
Gorelick
N.
,
Wilson
D.R.
,
Kozielski
K.
et al.
(
2020
)
Nonviral polymeric nanoparticles for gene therapy in pediatric CNS malignancies
.
Nanomed. Nanotechnol. Biol. Med.
23
,
102115
154.
Conde
J.
,
Tian
F.
,
Hernandez
Y.
,
Bao
C.
,
Baptista
P.V.
,
Cui
D.
et al.
(
2015
)
RNAi-based glyconanoparticles trigger apoptotic pathways for in vitro and in vivo enhanced cancer-cell killing
.
Nanoscale
7
,
9083
9091
[PubMed]
155.
Muthiah
M.
,
Che
H.-L.
,
Kalash
S.
,
Jo
J.
,
Choi
S.-Y.
,
Kim
W.J.
et al.
(
2015
)
Formulation of glutathione responsive anti-proliferative nanoparticles from thiolated Akt1 siRNA and disulfide-crosslinked PEI for efficient anti-cancer gene therapy
.
Colloids Surf. B
126
,
322
327
156.
Cohen
Z.R.
,
Ramishetti
S.
,
Peshes-Yaloz
N.
,
Goldsmith
M.
,
Wohl
A.
,
Zibly
Z.
et al.
(
2015
)
Localized RNAi therapeutics of chemoresistant grade IV glioma using hyaluronan-grafted lipid-based nanoparticles
.
ACS Nano
9
,
1581
1591
[PubMed]
157.
Li
L.
,
Wang
R.
,
Wilcox
D.
,
Sarthy
A.
,
Lin
X.
,
Huang
X.
et al.
(
2013
)
Developing lipid nanoparticle-based siRNA therapeutics for hepatocellular carcinoma using an integrated approach
.
Mol. Cancer Ther.
12
,
2308
2318
[PubMed]
158.
Prencipe
F.
,
Diaferia
C.
,
Rossi
F.
,
Ronga
L.
and
Tesauro
D.
(
2021
)
Forward Precision Medicine: Micelles for Active Targeting Driven by Peptides
.
Molecules
26
,
4049
[PubMed]
159.
Podesta
J.E.
,
Al‐Jamal
K.T.
,
Herrero
M.A.
,
Tian
B.
,
Ali‐Boucetta
H.
,
Hegde
V.
et al.
(
2009
)
Antitumor activity and prolonged survival by carbon‐nanotube‐mediated therapeutic siRNA silencing in a human lung xenograft model
.
Small
5
,
1176
1185
[PubMed]
160.
Kara
G.
,
Parlar
A.
,
Cakmak
M.C.
,
Cokol
M.
,
Denkbas
E.B.
and
Bakan
F.
(
2020
)
Silencing of survivin and cyclin B1 through siRNA-loaded arginine modified calcium phosphate nanoparticles for non-small-cell lung cancer therapy
.
Colloids Surf. B
196
,
111340
161.
Ghosh
R.
,
Singh
L.C.
,
Shohet
J.M.
and
Gunaratne
P.H.
(
2013
)
A gold nanoparticle platform for the delivery of functional microRNAs into cancer cells
.
Biomaterials
34
,
807
816
[PubMed]
162.
Devulapally
R.
,
Sekar
N.M.
,
Sekar
T.V.
,
Foygel
K.
,
Massoud
T.F.
,
Willmann
J.r.K.
et al.
(
2015
)
Polymer nanoparticles mediated codelivery of antimiR-10b and antimiR-21 for achieving triple negative breast cancer therapy
.
ACS Nano
9
,
2290
2302
[PubMed]
163.
Ebrahimian
M.
,
Taghavi
S.
,
Mokhtarzadeh
A.
,
Ramezani
M.
and
Hashemi
M.
(
2017
)
Co-delivery of doxorubicin encapsulated PLGA nanoparticles and Bcl-xL shRNA using alkyl-modified PEI into breast cancer cells
.
Appl. Biochem. Biotechnol.
183
,
126
136
[PubMed]
164.
Jiang
H.-L.
,
Xu
C.-X.
,
Kim
Y.-K.
,
Arote
R.
,
Jere
D.
,
Lim
H.-T.
et al.
(
2009
)
The suppression of lung tumorigenesis by aerosol-delivered folate–chitosan-graft-polyethylenimine/Akt1 shRNA complexes through the Akt signaling pathway
.
Biomaterials
30
,
5844
5852
[PubMed]
165.
Cole
G.
,
Ali
A.A.
,
McErlean
E.
,
Mulholland
E.J.
,
Short
A.
,
McCrudden
C.M.
et al.
(
2019
)
DNA vaccination via RALA nanoparticles in a microneedle delivery system induces a potent immune response against the endogenous prostate cancer stem cell antigen
.
Acta Biomater.
96
,
480
490
[PubMed]
166.
Oberli
M.A.
,
Reichmuth
A.M.
,
Dorkin
J.R.
,
Mitchell
M.J.
,
Fenton
O.S.
,
Jaklenec
A.
et al.
(
2017
)
Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy
.
Nano Lett.
17
,
1326
1335
[PubMed]
167.
Bijnsdorp
I.V.
,
Giovannetti
E.
and
Peters
G.J.
(
2011
)
Analysis of drug interactions
.
Cancer cell culture
, pp.
421
434
,
Springer
168.
Chou
T.-C.
and
Talalay
P.
(
1984
)
Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors
.
Adv. Enzyme. Regul.
22
,
27
55
[PubMed]
169.
Boshuizen
J.
and
Peeper
D.S.
(
2020
)
Rational cancer treatment combinations: an urgent clinical need
.
Mol. Cell
78
,
1002
1018
[PubMed]
170.
Mokhtari
R.B.
,
Homayouni
T.S.
,
Baluch
N.
,
Morgatskaya
E.
,
Kumar
S.
,
Das
B.
et al.
(
2017
)
Combination therapy in combating cancer
.
Oncotarget
8
,
38022
38043
[PubMed]
171.
Wang
H.
and
Huang
Y.
(
2020
)
Combination therapy based on nano codelivery for overcoming cancer drug resistance
.
Med. Drug Discovery
6
,
100024
172.
Ma
L.
,
Kohli
M.
and
Smith
A.
(
2013
)
Nanoparticles for combination drug therapy
.
ACS Nano
7
,
9518
9525
[PubMed]
173.
Gurunathan
S.
,
Kang
M.-H.
,
Qasim
M.
and
Kim
J.-H.
(
2018
)
Nanoparticle-mediated combination therapy: two-in-one approach for cancer
.
Int. J. Mol. Sci.
19
,
3264
174.
de Menezes
D.E.L.
,
Hudon
N.
,
McIntosh
N.
and
Mayer
L.D.
(
2000
)
Molecular and pharmacokinetic properties associated with the therapeutics of bcl-2 antisense oligonucleotide G3139 combined with free and liposomal doxorubicin
.
Clin. Cancer Res.
6
,
2891
2902
[PubMed]
175.
Cortes
J.
,
Estey
E.
,
O'Brien
S.
,
Giles
F.
,
Shen
Y.
,
Koller
C.
et al.
(
2001
)
High‐dose liposomal daunorubicin and high‐dose cytarabine combination in patients with refractory or relapsed acute myelogenous leukemia
.
Cancer
92
,
7
14
[PubMed]
176.
Pei
J.
,
Zhang
C.
,
Gokhale
P.C.
,
Rahman
A.
,
Dritschilo
A.
,
Ahmad
I.
et al.
(
2004
)
Combination with liposome-entrapped, ends-modified raf antisense oligonucleotide (LErafAON) improves the anti-tumor efficacies of cisplatin, epirubicin, mitoxantrone, docetaxel and gemcitabine
.
Anticancer Drugs
15
,
243
253
[PubMed]
177.
Pastorino
F.
,
Brignole
C.
,
Di Paolo
D.
,
Nico
B.
,
Pezzolo
A.
,
Marimpietri
D.
et al.
(
2006
)
Targeting liposomal chemotherapy via both tumor cell–specific and tumor vasculature–specific ligands potentiates therapeutic efficacy
.
Cancer Res.
66
,
10073
10082
[PubMed]
178.
Livi
L.
,
Meattini
I.
,
De Luca Cardillo
C.
,
Mangoni
M.
,
Greto
D.
,
Petrucci
A.
et al.
(
2009
)
Non-pegylated liposomal doxorubicin in combination with cyclophosphamide or docetaxel as first-line therapy in metastatic breast cancer: a retrospective analysis
.
Tumori J.
95
,
422
426
179.
Jiang
J.
,
Yang
S.-j.
,
Wang
J.-c.
,
Yang
L.-j.
,
Xu
Z.-z.
,
Yang
T.
et al.
(
2010
)
Sequential treatment of drug-resistant tumors with RGD-modified liposomes containing siRNA or doxorubicin
.
Eur. J. Pharm. Biopharm.
76
,
170
178
[PubMed]
180.
Chen
J.
,
Sun
X.
,
Shao
R.
,
Xu
Y.
,
Gao
J.
and
Liang
W.
(
2017
)
VEGF siRNA delivered by polycation liposome-encapsulated calcium phosphate nanoparticles for tumor angiogenesis inhibition in breast cancer
.
Int. J. Nanomed.
12
,
6075
181.
Bisht
S.
,
Mizuma
M.
,
Feldmann
G.
,
Ottenhof
N.A.
,
Hong
S.-M.
,
Pramanik
D.
et al.
(
2010
)
Systemic administration of polymeric nanoparticle-encapsulated curcumin (NanoCurc) blocks tumor growth and metastases in preclinical models of pancreatic cancer
.
Mol. Cancer Ther.
9
,
2255
2264
[PubMed]
182.
Kumar
P.
,
Wasim
L.
,
Chopra
M.
and
Chhikara
A.
(
2018
)
Co-delivery of vorinostat and etoposide via disulfide cross-linked biodegradable polymeric nanogels: synthesis, characterization, biodegradation, and anticancer activity
.
AAPS PharmSciTech.
19
,
634
647
[PubMed]
183.
Prylutska
S.
,
Grynyuk
I.
,
Matyshevska
O.
,
Prylutskyy
Y.
,
Evstigneev
M.
,
Scharff
P.
et al.
(
2014
)
C 60 fullerene as synergistic agent in tumor-inhibitory doxorubicin treatment
.
Drugs R&D
14
,
333
340
[PubMed]
184.
Zhang
L.
,
Lu
Z.
,
Zhao
Q.
,
Huang
J.
,
Shen
H.
and
Zhang
Z.
(
2011
)
Enhanced chemotherapy efficacy by sequential delivery of siRNA and anticancer drugs using PEI‐grafted graphene oxide
.
Small
7
,
460
464
[PubMed]
185.
Zhang
X.-F.
,
Yan
Q.
,
Shen
W.
and
Gurunathan
S.
(
2016
)
Trichostatin A enhances the apoptotic potential of palladium nanoparticles in human cervical cancer cells
.
Int. J. Mol. Sci.
17
,
1354
186.
Zhao
L.
,
Yuan
W.
,
Tham
H.P.
,
Chen
H.
,
Xing
P.
,
Xiang
H.
et al.
(
2017
)
Fast‐clearable nanocarriers conducting chemo/photothermal combination therapy to inhibit recurrence of malignant tumors
.
Small
13
,
1700963
187.
Mehrotra
N.
,
Kharbanda
S.
and
Singh
H.
(
2020
)
Peptide-based combination nanoformulations for cancer therapy
.
Nanomedicine
15
,
2201
2217
[PubMed]
188.
Mallick
A.M.
,
Chakraborty
K.
,
Biswas
A.
,
Jan
S.
,
Dutta
C.
,
Dey
S.
et al.
(
2019
)
Emerging Peptide-Based Technologies in Cancer Therapy
.
Unravelling Cancer Signaling Pathways: A Multidisciplinary Approach
, pp.
13
49
,
Springer
189.
Yu
W.
,
He
X.
,
Yang
Z.
,
Yang
X.
,
Xiao
W.
,
Liu
R.
et al.
(
2019
)
Sequentially responsive biomimetic nanoparticles with optimal size in combination with checkpoint blockade for cascade synergetic treatment of breast cancer and lung metastasis
.
Biomaterials
217
,
119309
[PubMed]
190.
Gupta
D.
,
Kumar
M.
,
Tyagi
P.
,
Kapoor
S.
,
Tyagi
A.
,
Barman
T.K.
et al.
(
2018
)
Concomitant delivery of paclitaxel and NuBCP-9 peptide for synergistic enhancement of cancer therapy
.
Nanomed. Nanotechnol. Biol. Med.
14
,
1301
1313
191.
Wang
W.
,
Suga
T.
,
Hagimori
M.
,
Kuroda
N.
,
Fuchigami
Y.
and
Kawakami
S.
(
2018
)
Investigation of intracellular delivery of NuBCP-9 by conjugation with oligoarginines peptides in MDA-MB-231 cells
.
Biol. Pharm. Bull.
41
,
1448
1455
[PubMed]
192.
Wang
Z.
,
Zhang
R.X.
,
Zhang
T.
,
He
C.
,
He
R.
,
Ju
X.
et al.
(
2018
)
In situ proapoptotic peptide-generating rapeseed protein-based nanocomplexes synergize chemotherapy for cathepsin-B overexpressing breast cancer
.
ACS Appl. Mater. Interfaces
10
,
41056
41069
193.
Yan
J.
,
He
W.
,
Yan
S.
,
Niu
F.
,
Liu
T.
,
Ma
B.
et al.
(
2018
)
Self-assembled peptide–lanthanide nanoclusters for safe tumor therapy: overcoming and utilizing biological barriers to peptide drug delivery
.
ACS Nano
12
,
2017
2026
[PubMed]
194.
Pazgier
M.
,
Liu
M.
,
Zou
G.
,
Yuan
W.
,
Li
C.
,
Li
C.
et al.
(
2009
)
Structural basis for high-affinity peptide inhibition of p53 interactions with MDM2 and MDMX
.
Proc. Natl. Acad. Sci.
106
,
4665
4670
195.
Conte
C.
,
Moret
F.
,
Esposito
D.
,
Dal Poggetto
G.
,
Avitabile
C.
,
Ungaro
F.
et al.
(
2019
)
Biodegradable nanoparticles exposing a short anti-FLT1 peptide as antiangiogenic platform to complement docetaxel anticancer activity
.
Mater. Sci. Eng. C
102
,
876
886
196.
Charbgoo
F.
,
Alibolandi
M.
,
Taghdisi
S.M.
,
Abnous
K.
,
Soltani
F.
and
Ramezani
M.
(
2018
)
MUC1 aptamer-targeted DNA micelles for dual tumor therapy using doxorubicin and KLA peptide
.
Nanomed. Nanotechnol. Biol. Med.
14
,
685
697
197.
Wang
H.
,
Wang
H.
,
Liang
J.
,
Jiang
Y.
,
Guo
Q.
,
Peng
H.
et al.
(
2014
)
Cell-penetrating apoptotic peptide/p53 DNA nanocomplex as adjuvant therapy for drug-resistant breast cancer
.
Mol. Pharm.
11
,
3352
3360
[PubMed]
198.
Lim
C.
,
Kang
J.K.
,
Won
W.R.
,
Park
J.Y.
,
Han
S.M.
,
Kim
J.C.
et al.
(
2019
)
Co-delivery of D-(KLAKLAK) 2 peptide and chlorin e6 using a liposomal complex for synergistic cancer therapy
.
Pharmaceutics
11
,
293
199.
Nishida
S.
,
Koido
S.
,
Takeda
Y.
,
Homma
S.
,
Komita
H.
,
Takahara
A.
et al.
(
2014
)
Wilms tumor gene (WT1) peptide–based cancer vaccine combined with gemcitabine for patients with advanced pancreatic cancer
.
J. Immunother.
37
,
105
[PubMed]
200.
Inoda
S.
,
Morita
R.
,
Hirohashi
Y.
,
Torigoe
T.
,
Asanuma
H.
,
Nakazawa
E.
et al.
(
2011
)
The feasibility of Cep55/c10orf3 derived peptide vaccine therapy for colorectal carcinoma
.
Exp. Mol. Pathol.
90
,
55
60
[PubMed]
201.
Hazama
S.
,
Nakamura
Y.
,
Takenouchi
H.
,
Suzuki
N.
,
Tsunedomi
R.
,
Inoue
Y.
et al.
(
2014
)
A phase I study of combination vaccine treatment of five therapeutic epitope-peptides for metastatic colorectal cancer; safety, immunological response, and clinical outcome
.
J. Transl. Med.
12
,
1
11
[PubMed]
202.
Okuno
K.
,
Sugiura
F.
,
Inoue
K.
and
Sukegawa
Y.
(
2014
)
Clinical trial of a 7-peptide cocktail vaccine with oral chemotherapy for patients with metastatic colorectal cancer
.
Anticancer Res.
34
,
3045
3052
[PubMed]
203.
Suzuki
N.
,
Hazama
S.
,
Ueno
T.
,
Matsui
H.
,
Shindo
Y.
,
Iida
M.
et al.
(
2014
)
A phase I clinical trial of vaccination with KIF20A-derived peptide in combination with gemcitabine for patients with advanced pancreatic cancer
.
J. Immunother.
37
,
36
[PubMed]
204.
Staff
C.
,
Mozaffari
F.
,
Frödin
J.-E.
,
Mellstedt
H.
and
Liljefors
M.
(
2014
)
Telomerase (GV1001) vaccination together with gemcitabine in advanced pancreatic cancer patients
.
Int. J. Oncol.
45
,
1293
1303
[PubMed]
205.
Clifton
G.T.
,
Peoples
G.E.
and
Mittendorf
E.A.
(
2016
)
The development and use of the E75 (HER2 369–377) peptide vaccine
.
Future Oncol.
12
,
1321
1329
[PubMed]
206.
Fukai
F.
,
Hasebe
S.
,
Ueki
M.
,
Mutoh
M.
,
Ohgi
C.
,
Takahashi
H.
et al.
(
1997
)
Identification of the anti-adhesive site buried within the heparin-binding domain of fibronectin
.
J. Biochem.
121
,
189
192
207.
Matsunaga
T.
,
Fukai
F.
,
Miura
S.
,
Nakane
Y.
,
Owaki
T.
,
Kodama
H.
et al.
(
2008
)
Combination therapy of an anticancer drug with the FNIII14 peptide of fibronectin effectively overcomes cell adhesion-mediated drug resistance of acute myelogenous leukemia
.
Leukemia
22
,
353
360
[PubMed]
208.
Cichoń
T.
,
Smolarczyk
R.
,
Matuszczak
S.
,
Barczyk
M.
,
Jarosz
M.
and
Szala
S.
(
2014
)
DK 6 L 9 peptide combination with IL-12 inhibits the recurrence of tumors in mice
.
Arch. Immunol. Ther. Exp. (Warsz.)
62
,
341
351
[PubMed]
209.
Masuzawa
T.
,
Fujiwara
Y.
,
Okada
K.
,
Nakamura
A.
,
Takiguchi
S.
,
Nakajima
K.
et al.
(
2012
)
Phase I/II study of S-1 plus cisplatin combined with peptide vaccines for human vascular endothelial growth factor receptor 1 and 2 in patients with advanced gastric cancer
.
Int. J. Oncol.
41
,
1297
1304
[PubMed]
210.
Chakraborty
K.
,
Dutta
C.
,
Mukherjee
S.
,
Biswas
A.
,
Gayen
P.
,
George
G.
et al.
(
2018
)
Engineering ionophore gramicidin‐inspired self‐assembled peptides for drug delivery and cancer nanotherapeutics
.
Adv. Therapeutics
1
,
1800018
211.
Lee
S.-M.
,
O'Halloran
T.V.
and
Nguyen
S.T.
(
2010
)
Polymer-caged nanobins for synergistic cisplatin− doxorubicin combination chemotherapy
.
J. Am. Chem. Soc.
132
,
17130
17138
[PubMed]
212.
Feldman
E.J.
,
Lancet
J.E.
,
Kolitz
J.E.
,
Ritchie
E.K.
,
Roboz
G.J.
,
List
A.F.
et al.
(
2011
)
First-in-man study of CPX-351: a liposomal carrier containing cytarabine and daunorubicin in a fixed 5: 1 molar ratio for the treatment of relapsed and refractory acute myeloid leukemia
.
J. Clin. Oncol.
29
,
979
[PubMed]
213.
Batist
G.
,
Gelmon
K.A.
,
Chi
K.N.
,
Miller
W.H.
,
Chia
S.K.
,
Mayer
L.D.
et al.
(
2009
)
Safety, pharmacokinetics, and efficacy of CPX-1 liposome injection in patients with advanced solid tumors
.
Clin. Cancer Res.
15
,
692
700
[PubMed]
214.
Dicko
A.
,
Frazier
A.A.
,
Liboiron
B.D.
,
Hinderliter
A.
,
Ellena
J.F.
,
Xie
X.
et al.
(
2008
)
Intra and inter-molecular interactions dictate the aggregation state of irinotecan co-encapsulated with floxuridine inside liposomes
.
Pharm. Res.
25
,
1702
1713
[PubMed]
215.
Wong
M.-Y.
and
Chiu
G.N.
(
2011
)
Liposome formulation of co-encapsulated vincristine and quercetin enhanced antitumor activity in a trastuzumab-insensitive breast tumor xenograft model
.
Nanomed. Nanotechnol. Biol. Med.
7
,
834
840
216.
Chen
Y.
,
Bathula
S.R.
,
Li
J.
and
Huang
L.
(
2010
)
Multifunctional nanoparticles delivering small interfering RNA and doxorubicin overcome drug resistance in cancer
.
J. Biol. Chem.
285
,
22639
22650
[PubMed]
217.
Agrawal
V.
,
Paul
M.K.
and
Mukhopadhyay
A.K.
(
2005
)
6-mercaptopurine and daunorubicin double drug liposomes—preparation, drug-drug interaction and characterization
.
J. Liposome Res.
15
,
141
155
[PubMed]
218.
Ren
Y.
,
Kang
C.-S.
,
Yuan
X.-B.
,
Zhou
X.
,
Xu
P.
,
Han
L.
et al.
(
2010
)
Co-delivery of as-miR-21 and 5-FU by poly (amidoamine) dendrimer attenuates human glioma cell growth in vitro
.
J. Biomater. Sci. Polym. Ed.
21
,
303
314
[PubMed]
219.
Lee
I.-H.
,
An
S.
,
Yu
M.K.
,
Kwon
H.-K.
,
Im
S.-H.
and
Jon
S.
(
2011
)
Targeted chemoimmunotherapy using drug-loaded aptamer–dendrimer bioconjugates
.
J. Control. Release
155
,
435
441
[PubMed]
220.
Clementi
C.
,
Miller
K.
,
Mero
A.
,
Satchi-Fainaro
R.
and
Pasut
G.
(
2011
)
Dendritic poly (ethylene glycol) bearing paclitaxel and alendronate for targeting bone neoplasms
.
Mol. Pharm.
8
,
1063
1072
[PubMed]
221.
Kaneshiro
T.L.
and
Lu
Z.-R.
(
2009
)
Targeted intracellular codelivery of chemotherapeutics and nucleic acid with a well-defined dendrimer-based nanoglobular carrier
.
Biomaterials
30
,
5660
5666
[PubMed]
222.
Tekade
R.K.
,
Dutta
T.
,
Tyagi
A.
,
Bharti
A.C.
,
Das
B.C.
and
Jain
N.K.
(
2008
)
Surface-engineered dendrimers for dual drug delivery: a receptor up-regulation and enhanced cancer targeting strategy
.
J. Drug Target.
16
,
758
772
[PubMed]
223.
Milane
L.
,
Duan
Z.-f.
and
Amiji
M.
(
2011
)
Pharmacokinetics and biodistribution of lonidamine/paclitaxel loaded, EGFR-targeted nanoparticles in an orthotopic animal model of multi-drug resistant breast cancer
.
Nanomed. Nanotechnol. Biol. Med.
7
,
435
444
224.
Shin
H.-C.
,
Alani
A.W.
,
Rao
D.A.
,
Rockich
N.C.
and
Kwon
G.S.
(
2009
)
Multi-drug loaded polymeric micelles for simultaneous delivery of poorly soluble anticancer drugs
.
J. Control. Release
140
,
294
300
[PubMed]
225.
Yang
T.
,
Wang
Y.
,
Li
Z.
,
Dai
W.
,
Yin
J.
,
Liang
L.
et al.
(
2012
)
Targeted delivery of a combination therapy consisting of combretastatin A4 and low-dose doxorubicin against tumor neovasculature
.
Nanomed. Nanotechnol. Biol. Med.
8
,
81
92
226.
Zhu
C.
,
Jung
S.
,
Luo
S.
,
Meng
F.
,
Zhu
X.
,
Park
T.G.
et al.
(
2010
)
Co-delivery of siRNA and paclitaxel into cancer cells by biodegradable cationic micelles based on PDMAEMA–PCL–PDMAEMA triblock copolymers
.
Biomaterials
31
,
2408
2416
[PubMed]
227.
Katragadda
U.
,
Teng
Q.
,
Rayaprolu
B.M.
,
Chandran
T.
and
Tan
C.
(
2011
)
Multi-drug delivery to tumor cells via micellar nanocarriers
.
Int. J. Pharm.
419
,
281
286
[PubMed]
228.
Wang
Y.
,
Gao
S.
,
Ye
W.-H.
,
Yoon
H.S.
and
Yang
Y.-Y.
(
2006
)
Co-delivery of drugs and DNA from cationic core–shell nanoparticles self-assembled from a biodegradable copolymer
.
Nat. Mater.
5
,
791
796
[PubMed]
229.
Bae
Y.
,
Diezi
T.A.
,
Zhao
A.
and
Kwon
G.S.
(
2007
)
Mixed polymeric micelles for combination cancer chemotherapy through the concurrent delivery of multiple chemotherapeutic agents
.
J. Control. Release
122
,
324
330
[PubMed]
230.
Wong
H.L.
,
Bendayan
R.
,
Rauth
A.M.
and
Wu
X.Y.
(
2006
)
Simultaneous delivery of doxorubicin and GG918 (Elacridar) by new Polymer-Lipid Hybrid Nanoparticles (PLN) for enhanced treatment of multidrug-resistant breast cancer
.
J. Control. Release
116
,
275
284
[PubMed]
231.
Song
X.R.
,
Zheng
Y.
,
He
G.
,
Yang
L.
,
Luo
Y.F.
,
He
Z.Y.
et al.
(
2010
)
Development of PLGA nanoparticles simultaneously loaded with vincristine and verapamil for treatment of hepatocellular carcinoma
.
J. Pharm. Sci.
99
,
4874
4879
[PubMed]
232.
Patil
Y.
,
Sadhukha
T.
,
Ma
L.
and
Panyam
J.
(
2009
)
Nanoparticle-mediated simultaneous and targeted delivery of paclitaxel and tariquidar overcomes tumor drug resistance
.
J. Control. Release
136
,
21
29
[PubMed]
233.
Djordjevic
J.
,
Del Rosario
L.S.
,
Wang
J.
and
Uhrich
K.E.
(
2008
)
Amphiphilic scorpion-like macromolecules as micellar nanocarriers
.
J. Bioactive Compatible Polymers
23
,
532
551
234.
Soma
C.E.
,
Dubernet
C.
,
Bentolila
D.
,
Benita
S.
and
Couvreur
P.
(
2000
)
Reversion of multidrug resistance by co-encapsulation of doxorubicin and cyclosporin A in polyalkylcyanoacrylate nanoparticles
.
Biomaterials
21
,
1
7
[PubMed]
235.
Surnar
B.
and
Jayakannan
M.
(
2016
)
Triple block nanocarrier platform for synergistic cancer therapy of antagonistic drugs
.
Biomacromolecules
17
,
4075
4085
[PubMed]
236.
Wang
X.
,
Yang
C.
,
Zhang
Y.
,
Zhen
X.
,
Wu
W.
and
Jiang
X.
(
2014
)
Delivery of platinum (IV) drug to subcutaneous tumor and lung metastasis using bradykinin-potentiating peptide-decorated chitosan nanoparticles
.
Biomaterials
35
,
6439
6453
[PubMed]
237.
Xiao
H.
,
Song
H.
,
Yang
Q.
,
Cai
H.
,
Qi
R.
,
Yan
L.
et al.
(
2012
)
A prodrug strategy to deliver cisplatin (IV) and paclitaxel in nanomicelles to improve efficacy and tolerance
.
Biomaterials
33
,
6507
6519
[PubMed]
238.
Zheng
C.
,
Zheng
M.
,
Gong
P.
,
Deng
J.
,
Yi
H.
,
Zhang
P.
et al.
(
2013
)
Polypeptide cationic micelles mediated co-delivery of docetaxel and siRNA for synergistic tumor therapy
.
Biomaterials
34
,
3431
3438
[PubMed]
239.
Zhu
C.
,
Xiao
J.
,
Tang
M.
,
Feng
H.
,
Chen
W.
and
Du
M.
(
2017
)
Platinum covalent shell cross-linked micelles designed to deliver doxorubicin for synergistic combination cancer therapy
.
Int. J. Nanomed.
12
,
3697
240.
Jang
Y.L.
,
Yun
U.J.
,
Lee
M.S.
,
Kim
M.G.
,
Son
S.
,
Lee
K.
et al.
(
2012
)
Cell-penetrating peptide mimicking polymer-based combined delivery of paclitaxel and siRNA for enhanced tumor growth suppression
.
Int. J. Pharm.
434
,
488
493
[PubMed]
241.
Salzano
G.
,
Navarro
G.
,
Trivedi
M.S.
,
De Rosa
G.
and
Torchilin
V.P.
(
2015
)
Multifunctional polymeric micelles co-loaded with anti–survivin siRNA and paclitaxel overcome drug resistance in an animal model of ovarian cancer
.
Mol. Cancer Ther.
14
,
1075
1084
[PubMed]
242.
Wang
N.
,
Wang
Z.
,
Nie
S.
,
Song
L.
,
He
T.
,
Yang
S.
et al.
(
2017
)
Biodegradable polymeric micelles coencapsulating paclitaxel and honokiol: a strategy for breast cancer therapy in vitro and in vivo
.
Int. J. Nanomed.
12
,
1499
243.
Ruttala
H.B.
,
Chitrapriya
N.
,
Kaliraj
K.
,
Ramasamy
T.
,
Shin
W.H.
,
Jeong
J.-H.
et al.
(
2017
)
Facile construction of bioreducible crosslinked polypeptide micelles for enhanced cancer combination therapy
.
Acta Biomater.
63
,
135
149
[PubMed]
244.
Krakovičová
H.
,
Etrych
T.
and
Ulbrich
K.
(
2009
)
HPMA-based polymer conjugates with drug combination
.
Eur. J. Pharm. Sci.
37
,
405
412
[PubMed]
245.
Segal
E.
,
Pan
H.
,
Ofek
P.
,
Udagawa
T.
,
Kopečková
P.
,
Kopeček
J.
et al.
(
2009
)
Targeting angiogenesis-dependent calcified neoplasms using combined polymer therapeutics
.
PLoS ONE
4
,
e5233
[PubMed]
246.
Pasut
G.
,
Greco
F.
,
Mero
A.
,
Mendichi
R.
,
Fante
C.
,
Green
R.J.
et al.
(
2009
)
Polymer−drug conjugates for combination anticancer therapy: investigating the mechanism of action
.
J. Med. Chem.
52
,
6499
6502
[PubMed]
247.
Santucci
L.
,
Mencarelli
A.
,
Renga
B.
,
Pasut
G.
,
Veronese
F.
,
Zacheo
A.
et al.
(
2006
)
Nitric oxide modulates proapoptotic and antiapoptotic properties of chemotherapy agents: the case of NO‐pegylated epirubicin
.
FASEB J.
20
,
765
767
[PubMed]
248.
Santucci
L.
,
Mencarelli
A.
,
Renga
B.
,
Ceccobelli
D.
,
Pasut
G.
,
Veronese
F.M.
et al.
(
2007
)
Cardiac safety and antitumoral activity of a new nitric oxide derivative of pegylated epirubicin in mice
.
Anticancer Drugs
18
,
1081
1091
[PubMed]
249.
Chandna
P.
,
Khandare
J.J.
,
Ber
E.
,
Rodriguez-Rodriguez
L.
and
Minko
T.
(
2010
)
Multifunctional tumor-targeted polymer-peptide-drug delivery system for treatment of primary and metastatic cancers
.
Pharm. Res.
27
,
2296
2306
[PubMed]
250.
Lee
J.H.
,
Sabnis
G.
,
Brodie
A.
and
Nan
A.
(
2011
)
In vitro evaluation of HER2 targeted N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer conjugates in combination treatment with tyrosine kinase inhibitor (PKI-166)
.
AACR
71
,
3230
251.
Lammers
T.
,
Subr
V.
,
Ulbrich
K.
,
Peschke
P.
,
Huber
P.E.
,
Hennink
W.E.
et al.
(
2009
)
Simultaneous delivery of doxorubicin and gemcitabine to tumors in vivo using prototypic polymeric drug carriers
.
Biomaterials
30
,
3466
3475
[PubMed]
252.
Xu
Q.
,
Leong
J.
,
Chua
Q.Y.
,
Chi
Y.T.
,
Chow
P.K.-H.
,
Pack
D.W.
et al.
(
2013
)
Combined modality doxorubicin-based chemotherapy and chitosan-mediated p53 gene therapy using double-walled microspheres for treatment of human hepatocellular carcinoma
.
Biomaterials
34
,
5149
5162
[PubMed]
253.
Lin
Y.-W.
,
Raj
E.N.
,
Liao
W.-S.
,
Lin
J.
,
Liu
K.-K.
,
Chen
T.-H.
et al.
(
2017
)
Co-delivery of paclitaxel and cetuximab by nanodiamond enhances mitotic catastrophe and tumor inhibition
.
Sci. Rep.
7
,
1
11
[PubMed]
254.
Thapa
R.K.
,
Byeon
J.H.
,
Choi
H.-G.
,
Yong
C.S.
and
Kim
J.O.
(
2017
)
PEGylated lipid bilayer-wrapped nano-graphene oxides for synergistic co-delivery of doxorubicin and rapamycin to prevent drug resistance in cancers
.
Nanotechnology
28
,
295101
[PubMed]
255.
Nair
G.G.
and
Nair
C.K.K.
(
2014
)
Sanazole directed targeting of silver nanoparticle drug complex to tumor mass: A preclinical investigation in murine model
.
J. Cancer Res. Therapeut.
10
,
979
256.
Tomuleasa
C.
,
Soritau
O.
,
Orza
A.
,
Dudea
M.
,
Petrushev
B.
,
Mosteanu
O.
et al.
(
2012
)
Gold nanoparticles conjugated with cisplatin/doxorubicin/capecitabine lower the chemoresistance of hepatocellular carcinoma-derived cancer cells
.
J. Gastrointestinal Liver Dis.
21
,
187
196
257.
Tolcher
A.W.
and
Mayer
L.D.
(
2018
)
Improving combination cancer therapy: the CombiPlex® development platform
.
Fut. Oncol.
14
,
1317
1332
[PubMed]
258.
Zhang
R.X.
,
Wong
H.L.
,
Xue
H.Y.
,
Eoh
J.Y.
and
Wu
X.Y.
(
2016
)
Nanomedicine of synergistic drug combinations for cancer therapy–Strategies and perspectives
.
J. Control. Release
240
,
489
503
[PubMed]
259.
Abraham
S.A.
,
McKenzie
C.
,
Masin
D.
,
Ng
R.
,
Harasym
T.O.
,
Mayer
L.D.
et al.
(
2004
)
In vitro and in vivo characterization of doxorubicin and vincristine coencapsulated within liposomes through use of transition metal ion complexation and pH gradient loading
.
Clin. Cancer Res.
10
,
728
738
[PubMed]
260.
Aryal
S.
,
Hu
C.M.J.
and
Zhang
L.
(
2010
)
Combinatorial drug conjugation enables nanoparticle dual‐drug delivery
.
Small
6
,
1442
1448
[PubMed]
261.
Shyam Matlapudi
M.
,
Moin
A.
,
Medishetti
R.
and
Rajendra
K.
(
2015
)
M Raichur, A.; Prashantha Kumar, B., Dual Drug Conjugate Loaded Nanoparticles for the Treatment of Cancer
.
Curr. Drug Deliv.
12
,
782
794
262.
Yao
Y.
,
Zhou
Y.
,
Liu
L.
,
Xu
Y.
,
Chen
Q.
,
Wang
Y.
et al.
(
2020
)
Nanoparticle-based drug delivery in cancer therapy and its role in overcoming drug resistance
.
Front. Mol. Biosci.
7
,
193
[PubMed]
263.
Kummar
S.
,
Chen
H.X.
,
Wright
J.
,
Holbeck
S.
,
Millin
M.D.
,
Tomaszewski
J.
et al.
(
2010
)
Utilizing targeted cancer therapeutic agents in combination: novel approaches and urgent requirements
.
Nat. Rev. Drug Discov.
9
,
843
856
264.
Saputra
E.C.
,
Huang
L.
,
Chen
Y.
and
Tucker-Kellogg
L.
(
2018
)
Combination therapy and the evolution of resistance: the theoretical merits of synergism and antagonism in cancer
.
Cancer Res.
78
,
2419
2431
[PubMed]
265.
Palmer
A.C.
and
Sorger
P.K.
(
2017
)
Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy
.
Cell
171
,
1678.e13
1691.e13
[PubMed]
266.
Doroshow
J.H.
and
Simon
R.M.
(
2017
)
On the design of combination cancer therapy
.
Cell
171
,
1476
1478
[PubMed]
267.
Zacharakis
N.
,
Chinnasamy
H.
,
Black
M.
,
Xu
H.
,
Lu
Y.-C.
,
Zheng
Z.
et al.
(
2018
)
Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer
.
Nat. Med.
24
,
724
730
[PubMed]
268.
Liu
H.
,
Zhao
Z.
,
Zhang
L.
,
Li
Y.
,
Jain
A.
,
Barve
A.
et al.
(
2019
)
Discovery of low-molecular weight anti-PD-L1 peptides for cancer immunotherapy
.
J. Immunotherapy Cancer
7
,
1
14
269.
Yin
H.
,
Zhou
X.
,
Huang
Y.-H.
,
King
G.J.
,
Collins
B.M.
,
Gao
Y.
et al.
(
2021
)
Rational design of potent peptide inhibitors of the PD-1: PD-L1 interaction for cancer immunotherapy
.
J. Am. Chem. Soc.
143
,
18536
18547
[PubMed]
270.
Hazama
D.
,
Yin
Y.
,
Murata
Y.
,
Matsuda
M.
,
Okamoto
T.
,
Tanaka
D.
et al.
(
2020
)
Macrocyclic peptide-mediated blockade of the CD47-SIRPα interaction as a potential cancer immunotherapy
.
Cell Chem. Biol.
27
,
1181.e7
1191.e7
[PubMed]
271.
Malonis
R.J.
,
Lai
J.R.
and
Vergnolle
O.
(
2019
)
Peptide-based vaccines: current progress and future challenges
.
Chem. Rev.
120
,
3210
3229
[PubMed]
272.
Mittendorf
E.A.
,
Storrer
C.E.
,
Foley
R.J.
,
Harris
K.
,
Jama
Y.
,
Shriver
C.D.
et al.
(
2006
)
Evaluation of the HER2/neu‐derived peptide GP2 for use in a peptide‐based breast cancer vaccine trial
.
Cancer
106
,
2309
2317
[PubMed]
273.
Kuai
R.
,
Yuan
W.
,
Son
S.
,
Nam
J.
,
Xu
Y.
,
Fan
Y.
et al.
(
2018
)
Elimination of established tumors with nanodisc-based combination chemoimmunotherapy
.
Sci. Adv.
4
,
eaao1736
[PubMed]
274.
Seth
A.
,
Heo
M.B.
and
Lim
Y.T.
(
2014
)
Poly (γ-glutamic acid) based combination of water-insoluble paclitaxel and TLR7 agonist for chemo-immunotherapy
.
Biomaterials
35
,
7992
8001
[PubMed]
275.
Roy
A.
,
Singh
M.S.
,
Upadhyay
P.
and
Bhaskar
S.
(
2013
)
Nanoparticle mediated co-delivery of paclitaxel and a TLR-4 agonist results in tumor regression and enhanced immune response in the tumor microenvironment of a mouse model
.
Int. J. Pharm.
445
,
171
180
[PubMed]
276.
Heo
M.B.
,
Kim
S.-Y.
,
Yun
W.S.
and
Lim
Y.T.
(
2015
)
Sequential delivery of an anticancer drug and combined immunomodulatory nanoparticles for efficient chemoimmunotherapy
.
Int. J. Nanomed.
10
,
5981
277.
Lu
Y.
,
Wang
Y.
,
Miao
L.
,
Haynes
M.
,
Xiang
G.
and
Huang
L.
(
2016
)
Exploiting in situ antigen generation and immune modulation to enhance chemotherapy response in advanced melanoma: a combination nanomedicine approach
.
Cancer Lett.
379
,
32
38
[PubMed]
278.
Fan
Y.
,
Kuai
R.
,
Xu
Y.
,
Ochyl
L.J.
,
Irvine
D.J.
and
Moon
J.J.
(
2017
)
Immunogenic cell death amplified by co-localized adjuvant delivery for cancer immunotherapy
.
Nano Lett.
17
,
7387
7393
[PubMed]
279.
Makkouk
A.
,
Joshi
V.B.
,
Wongrakpanich
A.
,
Lemke
C.D.
,
Gross
B.P.
,
Salem
A.K.
et al.
(
2015
)
Biodegradable microparticles loaded with doxorubicin and CpG ODN for in situ immunization against cancer
.
AAPS J.
17
,
184
193
[PubMed]
280.
Yin
Y.
,
Hu
Q.
,
Xu
C.
,
Qiao
Q.
,
Qin
X.
,
Song
Q.
et al.
(
2018
)
Co-delivery of doxorubicin and interferon-γ by thermosensitive nanoparticles for cancer immunochemotherapy
.
Mol. Pharm.
15
,
4161
4172
[PubMed]
281.
Song
Q.
,
Yin
Y.
,
Shang
L.
,
Wu
T.
,
Zhang
D.
,
Kong
M.
et al.
(
2017
)
Tumor microenvironment responsive nanogel for the combinatorial antitumor effect of chemotherapy and immunotherapy
.
Nano Lett.
17
,
6366
6375
[PubMed]
282.
Kong
M.
,
Tang
J.
,
Qiao
Q.
,
Wu
T.
,
Qi
Y.
,
Tan
S.
et al.
(
2017
)
Biodegradable hollow mesoporous silica nanoparticles for regulating tumor microenvironment and enhancing antitumor efficiency
.
Theranostics
7
,
3276
[PubMed]
283.
Heo
M.B.
and
Lim
Y.T.
(
2014
)
Programmed nanoparticles for combined immunomodulation, antigen presentation and tracking of immunotherapeutic cells
.
Biomaterials
35
,
590
600
[PubMed]
284.
Pradhan
P.
,
Qin
H.
,
Leleux
J.A.
,
Gwak
D.
,
Sakamaki
I.
,
Kwak
L.W.
et al.
(
2014
)
The effect of combined IL10 siRNA and CpG ODN as pathogen-mimicking microparticles on Th1/Th2 cytokine balance in dendritic cells and protective immunity against B cell lymphoma
.
Biomaterials
35
,
5491
5504
[PubMed]
285.
Xu
Z.
,
Wang
Y.
,
Zhang
L.
and
Huang
L.
(
2014
)
Nanoparticle-delivered transforming growth factor-β siRNA enhances vaccination against advanced melanoma by modifying tumor microenvironment
.
ACS Nano
8
,
3636
3645
[PubMed]
286.
Ahmed
M.
,
Kumar
G.
,
Navarro
G.
,
Wang
Y.
,
Gourevitch
S.
,
Moussa
M.H.
et al.
(
2015
)
Systemic siRNA nanoparticle-based drugs combined with radiofrequency ablation for cancer therapy
.
PLoS ONE
10
,
e0128910
[PubMed]
287.
Wang
D.
,
Wang
T.
,
Liu
J.
,
Yu
H.
,
Jiao
S.
,
Feng
B.
et al.
(
2016
)
Acid-activatable versatile micelleplexes for PD-L1 blockade-enhanced cancer photodynamic immunotherapy
.
Nano Lett.
16
,
5503
5513
[PubMed]
288.
Guo
L.
,
Yan
D.D.
,
Yang
D.
,
Li
Y.
,
Wang
X.
,
Zalewski
O.
et al.
(
2014
)
Combinatorial photothermal and immuno cancer therapy using chitosan-coated hollow copper sulfide nanoparticles
.
ACS Nano
8
,
5670
5681
[PubMed]
289.
Bear
A.S.
,
Kennedy
L.C.
,
Young
J.K.
,
Perna
S.K.
,
Mattos Almeida
J.P.
et al.
(
2013
)
Elimination of metastatic melanoma using gold nanoshell-enabled photothermal therapy and adoptive T cell transfer
.
PLoS ONE
8
,
e69073
[PubMed]
290.
Wang
C.
,
Xu
L.
,
Liang
C.
,
Xiang
J.
,
Peng
R.
and
Liu
Z.
(
2014
)
Immunological responses triggered by photothermal therapy with carbon nanotubes in combination with anti‐CTLA‐4 therapy to inhibit cancer metastasis
.
Adv. Mater.
26
,
8154
8162
[PubMed]
291.
Liu
Y.
,
Maccarini
P.
,
Palmer
G.M.
,
Etienne
W.
,
Zhao
Y.
,
Lee
C.-T.
et al.
(
2017
)
Synergistic immuno photothermal nanotherapy (SYMPHONY) for the treatment of unresectable and metastatic cancers
.
Sci. Rep.
7
,
1
6
[PubMed]
292.
He
C.
,
Duan
X.
,
Guo
N.
,
Chan
C.
,
Poon
C.
,
Weichselbaum
R.R.
et al.
(
2016
)
Core-shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy
.
Nat. Commun.
7
,
1
12
293.
Duan
X.
,
Chan
C.
,
Guo
N.
,
Han
W.
,
Weichselbaum
R.R.
and
Lin
W.
(
2016
)
Photodynamic therapy mediated by nontoxic core–shell nanoparticles synergizes with immune checkpoint blockade to elicit antitumor immunity and antimetastatic effect on breast cancer
.
J. Am. Chem. Soc.
138
,
16686
16695
[PubMed]
294.
Xu
J.
,
Xu
L.
,
Wang
C.
,
Yang
R.
,
Zhuang
Q.
,
Han
X.
et al.
(
2017
)
Near-infrared-triggered photodynamic therapy with multitasking upconversion nanoparticles in combination with checkpoint blockade for immunotherapy of colorectal cancer
.
ACS Nano
11
,
4463
4474
[PubMed]
295.
Lu
K.
,
He
C.
,
Guo
N.
,
Chan
C.
,
Ni
K.
,
Weichselbaum
R.R.
et al.
(
2016
)
Chlorin-based nanoscale metal–organic framework systemically rejects colorectal cancers via synergistic photodynamic therapy and checkpoint blockade immunotherapy
.
J. Am. Chem. Soc.
138
,
12502
12510
[PubMed]
296.
Patel
R.
,
Czapar
A.E.
,
Fiering
S.
,
Oleinick
N.L.
and
Steinmetz
N.F.
(
2018
)
Radiation therapy combined with cowpea mosaic virus nanoparticle in situ vaccination initiates immune-mediated tumor regression
.
ACS Omega
3
,
3702
3707
[PubMed]
297.
Lu
K.
,
He
C.
,
Guo
N.
,
Chan
C.
,
Ni
K.
,
Lan
G.
et al.
(
2018
)
Low-dose X-ray radiotherapy–radiodynamic therapy via nanoscale metal–organic frameworks enhances checkpoint blockade immunotherapy
.
Nat. Biomed. Eng.
2
,
600
610
[PubMed]
298.
Schmid
P.
,
Adams
S.
,
Rugo
H.S.
,
Schneeweiss
A.
,
Barrios
C.H.
,
Iwata
H.
et al.
(
2018
)
Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer
.
N. Engl. J. Med.
379
,
2108
2121
[PubMed]
299.
Whitehead
K.A.
,
Langer
R.
and
Anderson
D.G.
(
2009
)
Knocking down barriers: advances in siRNA delivery
.
Nat. Rev. Drug Discovery
8
,
129
138
300.
Van den Bergh
J.M.
,
Smits
E.L.
,
Berneman
Z.N.
,
Hutten
T.J.
,
De Reu
H.
et al.
(
2017
)
Monocyte-derived dendritic cells with silenced PD-1 ligands and transpresenting interleukin-15 stimulate strong tumor-reactive T-cell expansion
.
Cancer Immunol. Res.
5
,
710
715
[PubMed]
301.
Chatterjee
D.K.
,
Fong
L.S.
and
Zhang
Y.
(
2008
)
Nanoparticles in photodynamic therapy: an emerging paradigm
.
Adv. Drug. Deliv. Rev.
60
,
1627
1637
[PubMed]
302.
Mroz
P.
and
Hamblin
M.R.
(
2011
)
The immunosuppressive side of PDT
.
Photochem. Photobiol. Sci.
10
,
751
758
303.
Sanche
L.
(
2009
)
Beyond radical thinking
.
Nature
461
,
358
359
[PubMed]
304.
Kachikwu
E.L.
,
Iwamoto
K.S.
,
Liao
Y.-P.
,
DeMarco
J.J.
,
Agazaryan
N.
,
Economou
J.S.
et al.
(
2011
)
Radiation enhances regulatory T cell representation
.
Int. J. Radiation Oncol.* Biol.* Physics
81
,
1128
1135
305.
Ngiow
S.F.
,
McArthur
G.A.
and
Smyth
M.J.
(
2015
)
Radiotherapy complements immune checkpoint blockade
.
Cancer Cell
27
,
437
438
[PubMed]
306.
Kulkarni
A.
,
Chandrasekar
V.
,
Natarajan
S.K.
,
Ramesh
A.
,
Pandey
P.
,
Nirgud
J.
et al.
(
2018
)
A designer self-assembled supramolecule amplifies macrophage immune responses against aggressive cancer
.
Nat. Biomed. Eng.
2
,
589
599
[PubMed]
307.
Shields
C.W.
,
Evans
M.A.
,
Wang
L.L.-W.
,
Baugh
N.
,
Iyer
S.
,
Wu
D.
et al.
(
2020
)
Cellular backpacks for macrophage immunotherapy
.
Sci. Adv.
6
,
eaaz6579
[PubMed]
308.
McArthur
G.A.
and
Ribas
A.
(
2013
)
Targeting oncogenic drivers and the immune system in melanoma
.
J. Clin. Oncol.
31
,
499
506
[PubMed]
309.
Atefi
M.
,
Avramis
E.
,
Lassen
A.
,
Wong
D.J.
,
Robert
L.
,
Foulad
D.
et al.
(
2014
)
Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma
.
Clin. Cancer Res.
20
,
3446
3457
[PubMed]
310.
Li
H.
,
Xiao
Y.
,
Li
Q.
,
Yao
J.
,
Yuan
X.
,
Zhang
Y.
et al.
(
2021
)
The allergy mediator histamine confers resistance to immunotherapy in cancer patients via activation of the macrophage histamine receptor H1
.
Cancer Cell
40
,
36e9
52e9
311.
Chatterjee
S.
,
Azad
B.B.
and
Nimmagadda
S.
(
2014
)
The intricate role of CXCR4 in cancer
.
Adv. Cancer. Res.
124
,
31
82
[PubMed]
312.
Feig
C.
,
Jones
J.O.
,
Kraman
M.
,
Wells
R.J.
,
Deonarine
A.
,
Chan
D.S.
et al.
(
2013
)
Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti–PD-L1 immunotherapy in pancreatic cancer
.
Proc. Natl. Acad. Sci.
110
,
20212
20217
313.
Villanueva
L.
,
Álvarez-Errico
D.
and
Esteller
M.
(
2020
)
The contribution of epigenetics to cancer immunotherapy
.
Trends Immunol.
41
,
676
691
[PubMed]
314.
Covre
A.
,
Coral
S.
,
Nicolay
H.
,
Parisi
G.
,
Fazio
C.
,
Colizzi
F.
et al.
(
2015
)
Antitumor activity of epigenetic immunomodulation combined with CTLA-4 blockade in syngeneic mouse models
.
Oncoimmunology
4
,
e1019978
[PubMed]
315.
Fang
H.
,
Guo
Z.
,
Chen
J.
,
Lin
L.
,
Hu
Y.
,
Li
Y.
et al.
(
2021
)
Combination of epigenetic regulation with gene therapy-mediated immune checkpoint blockade induces anti-tumour effects and immune response in vivo
.
Nat. Commun.
12
,
1
19
[PubMed]
316.
Hegde
P.S.
and
Chen
D.S.
(
2020
)
Top 10 challenges in cancer immunotherapy
.
Immunity
52
,
17
35
[PubMed]
317.
Bouchlaka
M.N.
(
2011
)
Cancer immunotherapy: limitations and potential strategies
.
Doctoral dissertation
http://hdl.handle.net/11714/3978
318.
Huber
V.
,
Camisaschi
C.
,
Berzi
A.
,
Ferro
S.
,
Lugini
L.
and
Triulzi
T.
(
2017
)
Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation
.
Seminars in cancer biology
Elsevier
,
74
89
.
et al. In
319.
Huang
R.-Y.
,
Francois
A.
,
McGray
A.R.
,
Miliotto
A.
and
Odunsi
K.
(
2017
)
Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer
.
Oncoimmunology
6
,
e1249561
[PubMed]
320.
Murciano-Goroff
Y.R.
,
Warner
A.B.
and
Wolchok
J.D.
(
2020
)
The future of cancer immunotherapy: microenvironment-targeting combinations
.
Cell Res.
30
,
507
519
[PubMed]

Author notes

*

These authors have contributed equally to this work.

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).