Combination Therapy Based On Nano Codelivery For Overcoming Cancer
Combination Therapy Based On Nano Codelivery For Overcoming Cancer
Combination Therapy Based On Nano Codelivery For Overcoming Cancer
Review Article
A R T I C L E I N F O A B S T R A C T
Article history: Multidrug resistance (MDR) has long been a major obstacle in the treatment of malignant tumors. Combination ther-
Received 12 November 2019 apy provides a viable and promising strategy to overcome MDR. This article discusses the advantages of nano-based
Received in revised form 26 December 2019 combination therapy and proposes a 3R delivery principle (right place, right time, and right dose) as a reference for
Accepted 21 March 2020
development of cancer nanomedicine containing drug combinations. The article also reviews the strategies of nano-
Available online 31 March 2020
based codelivery, with emphasis on the techniques designed to overcome chemoresistance, enhance drug targeting de-
livery, and reduce immunosuppression.
© 2020 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
http://dx.doi.org/10.1016/j.medidd.2020.100024
2590-0986/© 2020 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 1. Typical MDR mechanisms. Reprinted with permission from ref. [17].
mechanisms fall into either the cellular or non-cellular category, depending 3. Combination therapy to overcome tumor resistance
on whether they are governed by the internal or external environment of
cancer cells [14]. They may also be divided into pharmacokinetic and phar- 3.1. Why drug combination?
macodynamic resistance pathways [15]. The most common mechanisms of
tumor cell drug resistance include (1) decreased drug inflow, (2) increased Combination therapy have been shown to enhance treatment efficiency
drug efflux (related to drug efflux transporters), (3) enhanced DNA repair, in cancer patients, compared to monotherapy [43–45]. At optimum syner-
(4) altered drug-specific targets, and (5) changed apoptosis pathways gistic ratio, the reasonable combination of multiple drugs can significantly
[16], as illustrated in Fig. 1 [17]. For example, the up-regulation of efflux improve the therapeutic effect [46,47]. In addition to enhancing drug effi-
transporters after chemotherapy can actively remove their substrates from cacy, combination therapy reduces the therapeutic dose needed for treat-
tumor cells, leading to unmet intracellular drug concentrations and poor ment, minimizes the adverse reactions, and hinders the development of
therapeutic efficacy [18,19]. drug resistance via synergistic and complementary molecular mechanisms
Therapeutic strategies for targeting drug efflux, cancer stem cell (CSCs)- [48]. The development of combination cancer therapies (e.g., combined
mediated drug escape, abnormal tumor metabolism, immune suppression, chemo drugs, chemo and immunotherapies, chemo and targeted therapies,
epithelial-mesenchymal transition (EMT), and abnormal apoptosis have chemo and gene therapies, and targeted therapies and immunotherapies)
been proposed to overcome MDR. These strategies are summarized in requires a better understanding of tumor biology, molecular pathways,
Table 1. and the interactions between tumors and their microenvironments
Table 1
Various therapeutic strategies for overcoming tumor MDR.
Resistance mechanism Tumor Model Treatment strategy Ref
2
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 3. Schematic of different design strategies for the combination of two drugs with nanocarriers. Figure adapted from ref. [47].
3
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
used experimental animals in cancer research, typically show significant For a long time, nanomedicine has been regarded as the primary means
EPR effect, but large animals rarely do. Also, In a cancer study conducted of effectively delivering drugs to tumor cells via the so-called “passive” or
on dogs with spontaneous tumors, it was found that most carcinomas “active” targeting. However, no strong clinical data currently exists to sup-
contained considerable amounts of accumulated liposome, but the soft tis- port the claim of drug delivery to the right place. In addition to the failure of
sue sarcomas did not [58]. Therefore, it is believed that the EPR effect clinical trials, the unproductive R&D of nanomedicines has prompted scien-
largely depends on tumor histology, and that it is more likely to occur in tists to rethink the fact that “nanomedicine research has almost exclusively
fast-growing vascular tumors like carcinomas (leaky blood vessels) than focused on tumor targeted drug delivery” [63]. Consequently, the focus of
in slow-growing non-vascular tumors such as sarcomas [59]. cancer medicine research may shift from targeted drug delivery to combi-
So far, there has been little evidence of the EPR effect in human patients. nation therapy, particularly considering that the latter provides an ideal ap-
Commercially available nanomedicines, such as nab-paclitaxel and PEGylated plication of nanomedicines that have the unique advantage of achieving
liposomal doxorubicin (DOX), show little improvement in patients' overall sur- multiple drug codelivery. With the aid of nanocarriers, co-encapsulated
vival [60,61], which undermines the efficiency of nanomedicine in targeting drugs are delivered to the same “right place”, thereby laying the foundation
tumors. In fact, this efficiency is far lower than the value assumed by most re- for synergistic action. Prior to the development of nano-based codelivery
searchers, even in animal models. A meta-analysis of the published nanoparti- techniques, it was quite difficult to realizing a precise combinatory dose
cle delivery researches shows that the estimated median efficiency of delivery in the targeted tissues and cells. However, nano-based codelivery tech-
is only 0.7% of the injected nanoparticle dose [62]. niques can ensure the delivery of a precise dose ratio of drugs into tumor
Fig. 5. (A) Plasma drug concentration after intravenous injection of cytarabine and daunorubicin in the form of nanoparticles or free drugs. (B) Survival rates of cytarabine
and daunorubicin in mice treated with different drug molar ratios. Reproduced with permission from refs. [53,54].
4
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 6. Strategies of functionalized liposome delivery for solid tumor therapy. (A) Conventional liposomes can reach tumor tissues through EPR effect. (B) PEGylated
liposomes can increase circulation time. (C) Functionalized liposomes modified with appropriate ligands can reach the target site and release the drug. (D) Responsive
liposomes activate drug release only under specific environmental conditions. Figure adapted from ref. [68].
cells. This is an especially advantageous characteristic, considering that The hypoxia, acidity, and upregulation of certain proteases in the TME
therapeutic efficiency strongly depends on the ratio of combined drugs. can promote MDR [73,74]. For example, an acidic TME induces high ex-
For example, the precise delivery of the liposomal combination Vyxeos® pression of hypoxia inducible factor 1 (HIF-1) and p-glycoproteins, leading
in the optimum cytarabine to daunorubicin drug ratio of 5 to 1 extends to chemotherapy resistance [75]. In addition, weak base drugs are neutral-
the survival rate three times more than the dose ratio of 1 to 1 (Fig. 5b). ized under acidic conditions, and they tend to accumulate in acidic organ-
Compared to free drugs, nanomedicines can extend drug half-life, elles (e.g., lysosomes), thereby failing to reach the target sites
thereby increasing drug accumulation in the tumor. More importantly, (e.g., nucleus). This is the reason why tumor cells are readily resistant to
the release of drugs from nano-carriers can be controlled. To minimize weakly alkaline chemo drugs [76,77]. Table 2 lists the cells in TME and
the undesirable side effects, the drugs should be timely released into the the factors that are associated with drug resistance.
tumor cells so as not leak into the circulation system. Another key advan-
tage of nanomedicines is that they can ensure a relatively identical pharma- 5. Overcoming chemoresistance via codelivery
cokinetic behavior of the co-encapsulated drugs. A typical example is the
cytarabine and daunorubicin drugs that exhibit similar pharmacokinetic 5.1. Codelivery of small drugs
profiles in liposomal combination Vyxeos®, but show great difference as
free drugs (Fig. 5a). Kang et al. developed a mannosylated liposomal codelivery system of
doxorubicin (DOX) and dihydroartemisinin (DHA) for the treatment of
3.3.2. Design of nano-based codelivery systems for cancer therapy drug-resistant colon cancer [78]. DHA, a derivative of artemisinin, is clini-
Stimuli-responsive “smart” systems have been extensively used to con- cally used as an effective antimalarial drug, and its potential for anticancer
trol the release of drugs from nanomedicines by endogenous or exogenous has been established. DHA in the codelivery system re-sensitized HCT8/
stimuli [64,65], such as changes in temperature, pH, and tumor-associated ADR tumor cells to DOX, thereby efficiently suppressing the drug-
enzyme expression [66,67]. Surface modification is another common resistant colon cancer via different MDR reversal mechanisms, such as pref-
method that facilitates intratumor drug accumulation and release. Fig. 6 erential nuclear accumulation, enhanced apoptosis, and the induction of
presents the delivery strategies of functionalized liposomes. autophagy. Moreover, the codelivered DOX/DHA system downregulated
Bcl-xl, an important signaling pathway associated with chemoresistance,
4. TME and drug resistance resulting in an anti-MDR effect. Interestingly, mannose receptors are
overexpressed in drug-resistant colon cancer cells (HCT8/ADR). Therefore,
TME refers to the internal and external environment in which tumor colon tumor codelivery could be efficiently achieved using a simple design
cells live and develop. This microenvironment is comprised of the tumor of mannosylated nanoparticles.
cells themselves, as well as fibroblasts, immune cells, stromal and intersti- Zhao et al. established a mannose modification strategy for the design of
tial cells, microvessels, and infiltrated biomolecules (e.g., cytokines, mannosylated albumin nanoparticles for codelivery of disulfiram/copper
chemokines, metabolites, and nutrients that support tumor growth) complexes (DSF/Cu2+) and regorafenib (Rego). The system served to over-
[69,70]. The TME constitutes a dual barrier against drug therapy. As a phys- come drug resistance in colon cancer by modulating the TME [79]. The
ical barrier, TME decreases drug intratumor penetration and diffusion. complexation of DSF, a drug that has been widely used over the past six de-
Meanwhile, as a biological barrier, it regulates genetic mutations, epige- cades to control alcoholism, with Cu2+, yields a cytotoxic product to the
netic changes, and subtype polarization via crosstalk networks, biomole- colon cancer cells. Meanwhile, Rego contributes to the anti-angiogenesis
cules, and extracellular vesicles, thereby inducing drug resistance. Further and repolarization of tumor-associated macrophages (TAM). As for albu-
details regarding TME and drug resistance are presented in Fig. 7 [71,72]. min, it is an important nutrient to cancer cells that are characterized by
5
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 7. Effect of treatment-activated TME on drug resistance. (A) Healthy person. (B) Diagnosis of solid tumor (e.g., lung adenocarcinoma). (C) Appearance of drug-resistant
phenotypes after treatment. (D) Association between mortality and tumor recurrence or metastasis. (E) Effect of combination therapies targeting both, cancer cells and TME
on treatment sensitivity and response. (F) Effect of combination therapy on treatment outcomes. Reproduced with permission from ref. [71].
overexpressed albumin-binding proteins (e.g., SPARC). Therefore, the use This enhanced the therapeutic effect of the drug codelivery system on
of albumin nanoparticles for codelivery promoted the uptake of the com- drug-resistant NSCLC.
bined DSF/Cu and Rego drugs by the cancer cells.
Jin et al. reported another codelivery system based on the activatable 5.2. Codelivery of small drugs and DNA
cell-penetrating peptide-modified liposomes. This system was composed
of simvastatin and paclitaxel, and used to overcome epithelial- Intrinsic apoptosis dysfunction is a common cause of
mesenchymal transition (EMT)-related resistance in non-small cell lung chemoresistance. The activation of TRAIL-mediated extrinsic path-
cancer (NSCLC) [30]. The cell-penetration ability of this delivery system ways could potentially induce apoptosis, resulting in the
could be conditionally blocked and reinstalled by protease cleavage in the resensitization of chemoresistant tumors. Xu et al. developed a nano
tumor, which allowed for the activation of intratumor penetration and in- system for the codelivery of TRAIL DNA and monensin [80]. This sys-
tracellular uptake. As a key driving factor of drug resistance, EMT is signif- tem was composed of low-molecular-weight PEI (LMW-PEI) and sulf-
icant in the drug-resistant A549/T cells. Simvastatin reverses EMT, possibly hydryl cyclodextrin moieties that were crosslinked by disulfide
via the mechanism of cholesterol metabolism (Fig. 8). In addition, the bonds; the cyclodextrin component also served as a carrier of hydro-
codelivery liposomes promoted the M2-to-M1 repolarization of TAM, phobic monensin, an antibiotic and potential antitumor drug, via
thereby reducing the secretion of TGF-β, the primary inducer of EMT. host-guest inclusion, thus forming a structure termed Moβ-CD-SSPEI
Table 2
A list of factors promoting therapeutic resistance in tumor cells. Reproduced with permission from ref. [72]. (More details are available in the reference).
Proteins or miRNAs Resistant to Molecular mechanism
6
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 8. Protease legumain-activatable liposomes can disrupt lipid rafts (cholesterol-rich domains) and suppress the integrin-β3 and FAK signaling pathways and re-sensitize
the drug-resistant cancer cells to paclitaxel. The liposomes are able to re-polarize TAM via cholesterol-associated LXR/ABCA1 regulation, resulting in the increased TNF-α and
attenuated TGF-β. This effect promotes the remodeling of the TME and suppresses EMT. Reprinted with permission from ref. [30].
Fig. 9. Preparation of the RGD-γ-PGA/Moβ-CD-SSPEIpDNA nanocomplex. The inter-molecular crosslink of LMW-PEI with cyclodextrin is achieved via disulfide bonds.
Subsequently, monensin is loaded onto cyclodextrin via guest-host complexation. Moβ-CD-SSPEIpDNA binds with DNA, thereby forming the bioreducible polyplex cores
that are later coated with γ-PGA or RGD-γ-PGA. Reprinted with permission from ref. [80].
7
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
(Fig. 9). The Moβ-CD-SSPEI bound to DNA, thereby forming the results indicated that this treatment strategy effectively overcame cancer
bioreducible polyplex cores that were subsequently coated with drug resistance (Fig. 10).
poly-γ-glutamic acid (γ-PGA) or RGD-γ-PGA via charge interaction,
forming the nanocomplex. This redox-sensitive gene delivery system
facilitated the disassembly of the crosslinked LMW-PEI in tumor 5.4. Codelivery of small drugs and proteins
cells and promoted the efficient release of TRAIL DNA and monensin.
As a death receptor (DR) agonist, monensin sensitized cancer cells to Tang et al. designed a nano-codelivery system capable of overcom-
TRAIL protein therapy by upregulating DR5. The anti-tumor efficacy ing MDR and metastasis. This system is composed of trichosanthin
of the proposed codelivery system was demonstrated in the drug- (TCS) protein and albendazole (ABZ) (Fig. 11) [82]. First, recombinant
resistant HCT8/ADR colon tumor model. TCS containing low-molecular-weight protamine, a cell-penetrating
peptide (CPP), was prepared (termed rTL). Then, ABZ was loaded onto
5.3. Codelivery of small drugs and peptides the negatively charged albumin-coated silver nanoparticles (ABZ@
BSA/Ag NP). The interaction between the oppositely charged compo-
To overcome doxorubicin resistance, Wang et al. proposed a novel DOX- nents resulted in the self-assembly of the rTL/ABZ@BSA/Ag NP
peptide conjugate self-assembly system characterized by tumor homing, codelivery system. In vitro studies showed that this system effectively
pH- and ultrasound-responsiveness [81]. In addition to DOX, the codelivery inhibited the proliferation of drug-resistant tumor cells (A549/T and
system consisted of ELP-C8 and LHRH. ELP-C8 is an elastin-like polypeptide HCT8/ADR) by destroying the cytoskeleton, blocking the cell cycle,
that carried DOX drug by conjugating to C8 peptide. Meanwhile, the LHRH downregulating P-gp, and promoting apoptosis. The in vivo results con-
peptide ligand is a luteinizing hormone that can specifically target the firmed that the rTL/ABZ@BSA/Ag NP codelivery system efficiently sup-
highly expressed receptors on tumor cells. The ELP-C8 and LHRH-ELP-C8 pressed tumor growth and lung metastasis in the mice with A549/T
were successfully expressed in Escherichia coli by genetic engineering, and drug-resistant subcutaneous tumors. Therefore, combinations of rTL
then purified and conjugated with DOX. High-intensity focused ultrasound and ABZ can be used as a potential therapeutic strategy to overcome
(HIFU) was used to further enhance intracellular drug delivery. In vivo MDR in tumors.
Fig. 10. Design of an LHRH-ELP-DOX codelivery system combined with HIFU-enhanced chemotherapy for the treatment of DOX-resistant tumors. (A) Synthesis route of Dox
nanoconjugates. (B) Acceleration of DOX release under acidic conditions (HIFU promotes the cleavage of the hydrazone bond between Dox and LHRH-ELP-C8).
(C) Ultrasound further enhances the efficiency of LHRH-ELP-DOX in treating DOX-resistant tumors. Reprinted with permission from ref. [81].
8
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 11. The schematic illustration of the rTL/ABZ@BSA/Ag NPs for combination therapy. Reprinted with permission from ref. [82].
6. Overcoming resistance to molecular-targeting therapy by mutations (EGRPT790M ) that are associated with gefitinib resistance.
Codelivery This is mainly due to the effect of the mutated methionine
(M) residue in blocking the interaction between gefitinib and the ac-
Molecular-targeting therapy has become the mainstream in NSCLC tive EGFR pocket [83]. Peng et al. proposed a novel HER-2 antibody
treatment. Gefitinib is the first approved epidermal growth factor recep- and mannose-modified liposomal codelivery system (tLGV) for the
tor (EGFR) tyrosine kinase inhibitor (TKI). This drug is applied in the treatment of EGRPT790M-mutated NSCLC. This system contains gefi-
treatment of metastatic NSCLC with EGFR mutations (i.e. exon 19 dele- tinib and vorinostat that modulated the redox microenvironment for
tions or exon 21 L858R substitutions). However, nearly all the patients promoting the degradation of EGRPT790M [84]. Vorinostat is a histone
receiving TKI therapy would develop drug resistance. In fact, approxi- deacetylase (HDAC) inhibitor that stimulated TAM repolarization. The
mately 50% of these patients eventually develop secondary T790M liposomal system repolarized the M2 phenotype to M1 and promoted
Fig. 12. A scheme of tLGV delivery and therapeutic mechanism for the treatment of EGFRT790M-positive NSCLC. Reprinted with permission from ref. [84].
9
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Fig. 13. Preparation of the mannose-modified lactoferrin nanoparticles and the antitumor mechanisms. (A) The mannose-modified lactoferrin nanoparticles are co-
loaded with shikonin and JQ1 by heat-driven self-assembly. (B) The system can minimize immunosuppression by inhibiting glycolysis, reducing the production of lactic
acid, and repolarizing TAM into M1 subtype. The system also induces ICD and promotes the infiltration of cytotoxic T lymphocytes into the tumors. Reprinted with
permission from ref. [95].
ROS generation in cancer cells, resulting in the modulation of the in- 7. Overcoming immunosuppression by codelivery
tracellular redox condition via the ROS/NOX3/MsrA axis. The redox
condition promoted the degradation of EGFRT790M, ultimately leading The occurrence and development of tumors are closely related to the
to the resensitization of resistant tumor cells to gefitinib (Fig. 12). body's immune system that can either suppress or promote tumor progres-
Yin et al. developed another liposomal codelivery system for the sion [86]. As they evolve, cancer cells recruit immunosuppressive cells and
treatment of EGFRT790M-positive NSCLC. They used a PD-L1 nanobody secrete soluble components to form an immunosuppressive network. This
as a targeting ligand for liposomal codelivery of simvastatin and gefi- leads to immune evasion and drug resistance, which ultimately promotes
tinib. This system could reverse gefitinib resistance by remodeling the the growth/progression of the tumor [87]. In general, immunosuppression
TME (e.g., neovascularization, macrophage polarization, and innate im- is closely related to a variety of immune cells (e.g., TAM and Tregs), as well
munity) [85]. as to some non-immune cells, such as fibroblasts and endothelial cells in
TME [88–90]. This indicates that the tumor immune microenvironment
10
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
Table 3
The latest combined drug therapy applications used to overcome drug resistance.
Drug delivery system Treatment strategy Payload Tumor type Ref
Mitochondria targeting nanoparticulate system Down-regulating pump resistance-related proteins P-gp siRNA and mitochondria MCF-7/ADR [97]
complex
Dual stimuli-responsive Pt(IV) prodrug Targeting NQO1 Three hybrid compounds A549 [98]
Synergistic therapy (CT, PDT, and PTT) Inhibiting drug efflux TPGS & DOX MCF-7/ADR [99]
NP − peptide−drug bioconjugate Improving drug delivery efficiency DOX peptides H69AR [100]
pH/Redox Dual-Responsive Polyplex Endo-lysosomal escape MDR1 siRNA & DOX MCF-7/ADR [101]
A folate-decorated polymersome Co-loaded chemotherapy drugs and P-gp inhibitors Tariquidar & DOX & paclitaxel MDR MCF-7 [102]
Antibody-drug conjugate c-Met-targeting c-Met mAb and microtubule HCC827 [103]
inhibitor
Combination EGFR+FGFR inhibitors Overcoming EMT Gefitinib and BGJ398 PC9 [104]
Drug delivery micelles Synergistic therapy of Image-guided PDT and CT PTX & tariquidar & fluorophore SKOV-3/MDR [105]
pH-and photothermal-driven multistage delivery Hyperthermia and chemotherapy DOX and Gold nanocages MCF-7/ADR [106]
nanoplatforms
Polymer-drug conjugates Bypassed drug efflux pumps DOX and MPP MCF-7/ADR [107]
Zinc oxide nanoparticle Increasing ROS generation synergistic autophagy Zinc oxide and DOX MCF-7/ADR [108]
CB-839 plus metformin Targeting glutamine-addiction CB-839 and metformin ESCC [109]
Implantable hierarchical-structured micelle−/drug-loaded Inhibiting drug efflux, increasing the intracellular DOX DOX and Apatinib MCF-7/ADR [110]
fiber device accumulation
Dual pH-sensitive liposomal system Charge reversal promotes drug release inhibition drug Paclitaxel and NO donor A549/T [111]
efflux
Liposome Sequential release promotes drug accumulation in tumor Dexamethasone (DEX) and KBv [112]
Docetaxel (DTX)
(TIME) is regulated by a highly complicated network and significantly af- reactions. Future nanomedicine research should address safety, biocompat-
fects the efficacy of immunotherapy [91]. ibility, availability, and toxicity issues. In addition, research efforts should
Tumor metabolism abnormality plays a key role in immunosuppression also focus on providing a profound understanding of the molecular regula-
and drug resistance [92,93]. For example, the Warburg effect, a hallmark of tory mechanism in tumors and TME. Such information is needed to design
cancer, is characterized by increased aerobic glycolysis, which enhances new drug delivery systems that are likely to achieve great success in cancer
the production of lactate, a known immunosuppressive metabolite in treatment.
TIME and an important driving factor of TAM2 polarization [94].
Wang et al. developed a mannose-modified lactoferrin nanosystem for Conflict of Interest
biomimetic delivery of shikonin (SHK) and JQ1. The system prepared via
heat-driven self-assembly served to reprogram the TIME and the glucose The authors declare that there are no conflicts of interest.
metabolism [95]. The reprogramming effect was achieved by targeting
the highly expressed mannose receptor and LRP-1 on tumor cells and Credit Author Statement
TAM, repolarizing TAM from M2 to M1 subtype, inducing immunogenic
tumor cell death (ICD), suppressing glycolysis, and promoting the H.W. and Y.H. wrote the manuscript.
intratumoral infiltration of CD8+ T cells. In addition, JQ1 reduced the ex-
pression of PD-L1 on tumor cells, thus, yielding a synergic effect and acti- Acknowledgments
vating anticancer immunity (Fig. 13).
We are thankful for the support of NFSC (81925035, 81673382, and
8. Summary of codelivery-based therapeutic applications 81521005), Strategic Priority Research Program of CAS (XDA12050307),
National Special Project for Significant New Drugs Development
Stimuli-sensitive nanosystems are responsive to the changes of pH, ROS, (2018ZX09711002-010-002), CAS Scientific Research and Equipment De-
or enzymes in TME. These systems are capable of releasing drugs rapidly velopment Project (YZ201437), and Fudan-SIMM Joint Research Fund
and efficiently in tumor cells. Li et al. provided a comprehensive summary (FU-SIMM20174009) for support.
of the nanometer drug delivery systems used to overcome MDR [96]. These
systems were based on extracellular (light or heat), intracellular (pH, ROS, References
reduction, or enzyme), or dual stimulation.
The tumor cell- and TME-targeting strategies for overcoming drug resis- [1] Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: good or bad
tance are illustrated in Fig. 14. Meanwhile, Table 3 provides a list of some news from the 2018 global cancer statistics? Cancer Commun (Lond) 2019;39:22.
combined drug therapy applications. Most contemporary methods focus [2] Gurunathan S, Kang MH, Qasim M, Kim JH. Nanoparticle-mediated combination ther-
apy: two-in-one approach for cancer. Int J Mol Sci 2018;19:pii: E3264.
on joint strategies to overcome drug resistance.
[3] Housman G, Byler S, Heerboth S, Lapinska K, Longacre M, Snyder N, et al. Drug resis-
tance in cancer: an overview. Cancers (Basel) 2014;6:1769–92.
9. Perspective [4] Steeg PS. Targeting metastasis. Nat Rev Cancer 2016;16:201–18.
[5] Mader RM, Schmidt WM, Steger GG, Krupitza G. Molecular mechanisms of drug resis-
tance. Int J Clin Pharmacol Ther 2009;47:49–50.
Despite the significant clinical breakthroughs witnessed in the field of
[6] Chen ZS, Tiwari AK. Multidrug resistance proteins (MRPs/ABCCs) in cancer chemo-
cancer therapy, tumor treatments are inevitably accompanied by drug resis- therapy and genetic diseases. FEBS J 2011;278:3226–45.
tance. Therefore, it has become essential to develop strategies for overcom- [7] Zhang M, Liu E, Cui Y, Huang Y. Nanotechnology-based combination therapy for over-
ing MDR. So far, nano-based drug combinations have shown great potential coming multidrug-resistant cancer. Cancer Biol Med 2017;14:212–27.
[8] Cho J, Chang YH, Heo YJ, Kim S, Kim NK, Park JO, et al. Four distinct immune micro-
for improving drug-resistant cancer therapy. In order to develop such
environment subtypes in gastric adenocarcinoma with special reference to microsatel-
nanomedicines, it is important to optimize the parameters of 3R delivery, lite instability. ESMO Open 2018;3:e000326.
as well as to assess the biological fates and molecular mechanisms of the [9] Chung HW, Lim JB. Role of the tumor microenvironment in the pathogenesis of gastric
combined drugs. carcinoma. World J Gastroenterol 2014;20:1667–80.
[10] Lane D. Designer combination therapy for cancer. Nat Biotechnol 2006;24:163–4.
Further research on nanomedicine is still needed to effectively over- [11] Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an
come drug resistance, improve patient survival, and reduce adverse evolving paradigm. Nat Rev Cancer 2013;13:714–26.
11
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
[12] Li YJ, Lei YH, Yao N, Wang CR, Hu N, Ye WC, et al. Autophagy and multidrug resis- [45] Jia J, Zhu F, Ma X, Cao Z, Cao ZW, Li Y, et al. Mechanisms of drug combinations: in-
tance in cancer. Chin J Cancer 2017;36:52. teraction and network perspectives. Nat Rev Drug Discov 2009;8:111–28.
[13] Chen S, Yang K, Tuguntaev RG, Mozhi A, Zhang J, Wang PC, et al. Targeting tumor mi- [46] Hu Q, Sun W, Wang C, Gu Z. Recent advances of cocktail chemotherapy by combina-
croenvironment with PEG-based amphiphilic nanoparticles to overcome tion drug delivery systems. Adv Drug Deliv Rev 2016;98:19–34.
chemoresistance. Nanomedicine 2016;12:269–86. [47] Zhang RX, Wong HL, Xue HY, Eoh JY, Wu XY. Nanomedicine of synergistic drug com-
[14] Kartal-Yandim M, Adan-Gokbulut A, Baran Y. Molecular mechanisms of drug resis- binations for cancer therapy - strategies and perspectives. J Control Release 2016;240:
tance and its reversal in cancer. Crit Rev Biotechnol 2016;36:716–26. 489–503.
[15] Mellor HR, Callaghan R. Resistance to chemotherapy in cancer: a complex and inte- [48] Chou TC. Theoretical basis, experimental design, and computerized simulation of syn-
grated cellular response. Pharmacology 2008;81:275–300. ergism and antagonism in drug combination studies. Pharmacol Rev 2006;58:621–81.
[16] Aleksakhina SN, Kashyap A, Imyanitov EN. Mechanisms of acquired tumor drug resis- [49] Sabnis AJ, Bivona TG. Principles of resistance to targeted Cancer therapy: lessons from
tance. Biochim Biophys Acta Rev Cancer 2019;1872:188310. basic and translational Cancer biology. Trends Mol Med 2019;25:185–97.
[17] Bar-Zeev M, Livney YD, Assaraf YG. Targeted nanomedicine for cancer therapeutics: to- [50] Wargo JA, Reuben A, Cooper ZA, Oh KS, Sullivan RJ. Immune effects of chemotherapy,
wards precision medicine overcoming drug resistance. Drug Resist Updat 2017;31:15–30. radiation, and targeted therapy and opportunities for combination with immunother-
[18] Harris AL, Hochhauser D. Mechanisms of multidrug resistance in cancer treatment. apy. Semin Oncol 2015;42:601–16.
Acta Oncol 1992;31:205–13. [51] Slovin S. Chemotherapy and immunotherapy combination in advanced prostate can-
[19] Kirtane AR, Kalscheuer SM, Panyam J. Exploiting nanotechnology to overcome tumor cer. Clin Adv Hematol Oncol 2012;10:90–100.
drug resistance: challenges and opportunities. Adv Drug Deliv Rev 2013;65:1731–47. [52] M. Bahri, J. Fleurence, S. Faraj, M. Ben Mostefa Daho, S. Fougeray, S. Birkle, Potenti-
[20] Wang H, Gao Z, Liu X, Agarwal P, Zhao S, Conroy DW, et al. Targeted production of ation of Anticancer Antibody Efficacy by Antineoplastic Drugs: Detection of
reactive oxygen species in mitochondria to overcome cancer drug resistance. Nat Antibody-drug Synergism Using the Combination Index Equation, J Vis Exp, (2019).
Commun 2018;9:562. [53] Ma L, Kohli M, Smith A. Nanoparticles for combination drug therapy. ACS Nano 2013;
[21] Christie EL, Pattnaik S, Beach J, Copeland A, Rashoo N, Fereday S, et al. Multiple 7:9518–25.
ABCB1 transcriptional fusions in drug resistant high-grade serous ovarian and breast [54] Tardi P, Johnstone S, Harasym N, Xie S, Harasym T, Zisman N, et al. In vivo mainte-
cancer. Nat Commun 2019;10:1295. nance of synergistic cytarabine:daunorubicin ratios greatly enhances therapeutic effi-
[22] Wang S, Xu Y, Chan HF, Kim HW, Wang Y, Leong KW, et al. Nanoparticle-mediated in- cacy. Leuk Res 2009;33:129–39.
hibition of survivin to overcome drug resistance in cancer therapy. J Control Release [55] Waterhouse DN, Gelmon KA, Klasa R, Chi K, Huntsman D, Ramsay E, et al. Develop-
2016;240:454–64. ment and assessment of conventional and targeted drug combinations for use in the
[23] Bugde P, Biswas R, Merien F, Lu J, Liu DX, Chen M, et al. The therapeutic potential of treatment of aggressive breast cancers. Curr Cancer Drug Targets 2006;6:455–89.
targeting ABC transporters to combat multi-drug resistance. Expert Opin Ther Targets [56] Livney YD, Assaraf YG. Rationally designed nanovehicles to overcome cancer
2017;21:511–30. chemoresistance. Adv Drug Deliv Rev 2013;65:1716–30.
[24] Wu Y, Zhang Y, Zhang W, Sun C, Wu J, Tang J. Reversing of multidrug resistance [57] Danhier F. To exploit the tumor microenvironment: since the EPR effect fails in the
breast cancer by co-delivery of P-gp siRNA and doxorubicin via folic acid-modified clinic, what is the future of nanomedicine? J Control Release 2016;244:108–21.
core-shell nanomicelles. Colloids Surf B Biointerfaces 2016;138:60–9. [58] A.E. Hansen, A.L. Petersen, J.R. Henriksen, B. Boerresen, P. Rasmussen, D.R. Elema, P.
[25] Lu X, Liu R, Wang M, Kumar AK, Pan F, He L, et al. MicroRNA-140 impedes DNA repair M. af Rosenschold, A.T. Kristensen, A. Kjaer, T.L. Andresen, Positron Emission Tomog-
by targeting FEN1 and enhances chemotherapeutic response in breast cancer. Onco- raphy Based Elucidation of the Enhanced Permeability and Retention Effect in Dogs
gene 2019;39:234–47. with Cancer Using Copper-64 Liposomes, ACS Nano, 9 (2015) 6985–6995.
[26] Fianco G, Contadini C, Ferri A, Cirotti C, Stagni V, Barila D. Caspase-8: a novel target to [59] A.Z. Wang, EPR or no EPR? The billion-dollar question, Science Translational Medi-
overcome resistance to chemotherapy in glioblastoma. Int J Mol Sci 2018;19 pii: cine, 7 (2015) 294ec112.
E3798. [60] T. Luhn, S. Chui, A. Hsieh, J. Yi, A. Mecke, P. Bajaj, W. Hasnain, A. Falgas, T.G.N. Ton,
[27] Tang Z, Takahashi Y, Chen C, Liu Y, He H, Tsotakos N, et al. Atg2A/B deficiency A. Kurian, 316PComparative effectiveness of nab-paclitaxel vs. paclitaxel monotherapy
switches cytoprotective autophagy to non-canonical caspase-8 activation and apopto- as first-line (1L) treatment of metastatic triple-negative breast cancer (mTNBC) in US
sis. Cell Death Differ 2017;24:2127–38. clinical practice, Annals of Oncology, 29 (2018).
[28] Fianco G, Mongiardi MP, Levi A, De Luca T, Desideri M, Trisciuoglio D, et al. Caspase-8 [61] O'Brien ME, Wigler N, Inbar M, Rosso R, Grischke E, Santoro A, et al. Reduced
contributes to angiogenesis and chemotherapy resistance in glioblastoma. Elife 2017;6. cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxo-
[29] Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting rubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment
tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosup- of metastatic breast cancer. Ann Oncol 2004;15:440–9.
pression, and improves chemotherapeutic responses. Cancer Res 2013;73:1128–41. [62] Wilhelm S, Tavares AJ, Dai Q, Ohta S, Audet J, Dvorak HF, et al. Analysis of nanopar-
[30] Jin H, He Y, Zhao P, Hu Y, Tao J, Chen J, et al. Targeting lipid metabolism to overcome ticle delivery to tumours. Nat Rev Mater 2016;1:16014.
EMT-associated drug resistance via integrin beta3/FAK pathway and tumor-associated [63] Park K. The beginning of the end of the nanomedicine hype. J Control Release 2019;
macrophage repolarization using legumain-activatable delivery. Theranostics 2019;9: 305:221–2.
265–78. [64] Siegel RA. Stimuli sensitive polymers and self regulated drug delivery systems: a very
[31] Shee K, Yang W, Hinds JW, Hampsch RA, Varn FS, Traphagen NA, et al. Therapeuti- partial review. J Control Release 2014;190:337–51.
cally targeting tumor microenvironment-mediated drug resistance in estrogen [65] Bhatnagar S, Venuganti VV. Cancer targeting: responsive polymers for stimuli-sensitive
receptor-positive breast cancer. J Exp Med 2018;215:895–910. drug delivery. J Nanosci Nanotechnol 2015;15:1925–45.
[32] Islam SS, Al-Sharif I, Sultan A, Al-Mazrou A, Remmal A, Aboussekhra A. Eugenol po- [66] Du Y, Chen B. Combination of drugs and carriers in drug delivery technology and its
tentiates cisplatin anti-cancer activity through inhibition of ALDH-positive breast can- development. Drug Des Devel Ther 2019;13:1401–8.
cer stem cells and the NF-kappaB signaling pathway. Mol Carcinog 2018;57:333–46. [67] Wu W, Luo L, Wang Y, Wu Q, Dai HB, Li JS, et al. Endogenous pH-responsive nanopar-
[33] Ojha R, Bhattacharyya S, Singh SK. Autophagy in cancer stem cells: a potential link be- ticles with programmable size changes for targeted tumor therapy and imaging appli-
tween chemoresistance, recurrence, and metastasis. Biores Open Access 2015;4:97–108. cations. Theranostics 2018;8:3038–58.
[34] Huang T, Song X, Yang Y, Wan X, Alvarez AA, Sastry N, et al. Autophagy and hallmarks [68] Riaz MK, Riaz MA, Zhang X, Lin C, Wong KH, Chen X, et al. Surface functionalization
of cancer. Crit Rev Oncog 2018;23:247–67. and targeting strategies of liposomes in solid tumor therapy: A review. Int J Mol Sci
[35] Ashkenazi A, Fairbrother WJ, Leverson JD, Souers AJ. From basic apoptosis discoveries 2018;19 pii: E195.
to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov 2017;16:273–84. [69] Wu T, Dai Y. Tumor microenvironment and therapeutic response. Cancer Lett 2017;
[36] Xu W, Tang W, Li T, Zhang X, Sun Y. Overcoming resistance to AC0010, a third gener- 387:61–8.
ation of EGFR inhibitor, by targeting c-MET and BCL-2. Neoplasia 2019;21:41–51. [70] Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates Cancer
[37] Zhang R, Wang SB, Chen AZ, Chen WG, Liu YG, Wu WG, et al. Codelivery of paclitaxel progression. Cancer Res 2019;79:4557–66.
and small interfering RNA by octadecyl quaternized carboxymethyl chitosan-modified [71] Sun Y. Tumor microenvironment and cancer therapy resistance. Cancer Lett 2016;380:
cationic liposome for combined cancer therapy. J Biomater Appl 2015;30:351–60. 205–15.
[38] Li Y, Luo J, Lin MT, Zhi P, Guo WW, Han M, et al. Co-delivery of metformin enhances [72] Son B, Lee S, Youn H, Kim E, Kim W, Youn B. The role of tumor microenvironment in
the antimultidrug resistant tumor effect of doxorubicin by improving hypoxic tumor therapeutic resistance. Oncotarget 2017;8:3933–45.
microenvironment. Mol Pharm 2019;16:2966–79. [73] Wojtkowiak JW, Verduzco D, Schramm KJ, Gillies RJ. Drug resistance and cellular ad-
[39] Li WQ, Wang Z, Hao S, He H, Wan Y, Zhu C, et al. Mitochondria-targeting aptation to tumor acidic pH microenvironment. Mol Pharm 2011;8:2032–8.
polydopamine nanoparticles to deliver doxorubicin for overcoming drug resistance. [74] Milane L, Duan Z, Amiji M. Role of hypoxia and glycolysis in the development of multi-
ACS Appl Mater Interfaces 2017;9:16793–802. drug resistance in human tumor cells and the establishment of an orthotopic multi-
[40] Y. Zhou, F. Huang, Y. Yang, P. Wang, Z. Zhang, Y. Tang, Y. Shen, K. Wang, Paraptosis- drug resistant tumor model in nude mice using hypoxic pre-conditioning. Cancer
inducing nanomedicine overcomes cancer drug resistance for a potent cancer therapy, Cell Int 2011;11:3.
Small, 14 (2018). [75] Tredan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor mi-
[41] Bosc C, Selak MA, Sarry JE. Resistance is futile: targeting mitochondrial energetics and croenvironment. J Natl Cancer Inst 2007;99:1441–54.
metabolism to overcome drug resistance in cancer treatment. Cell Metab 2017;26: [76] Das V, Stepankova J, Hajduch M, Miller JH. Role of tumor hypoxia in acquisition
705–7. of resistance to microtubule-stabilizing drugs. Biochim Biophys Acta 2015;1855:
[42] Yamaoka T, Ohba M, Ohmori T. Molecular-targeted therapies for epidermal growth 172–82.
factor receptor and its resistance mechanisms. Int J Mol Sci 2017;18 pii: E2420. [77] Taylor S, Spugnini EP, Assaraf YG, Azzarito T, Rauch C, Fais S. Microenvironment acid-
[43] Hu CM, Zhang L. Nanoparticle-based combination therapy toward overcoming drug re- ity as a major determinant of tumor chemoresistance: proton pump inhibitors (PPIs) as
sistance in cancer. Biochem Pharmacol 2012;83:1104–11. a novel therapeutic approach. Drug Resist Updat 2015;23:69–78.
[44] Valencia PM, Pridgen EM, Perea B, Gadde S, Sweeney C, Kantoff PW, et al. Synergistic [78] Kang XJ, Wang HY, Peng HG, Chen BF, Zhang WY, Wu AH, et al. Codelivery of
cytotoxicity of irinotecan and cisplatin in dual-drug targeted polymeric nanoparticles. dihydroartemisinin and doxorubicin in mannosylated liposomes for drug-resistant
Nanomedicine (Lond) 2013;8:687–98. colon cancer therapy. Acta Pharmacol Sin 2017;38:885–97.
12
H. Wang, Y. Huang / Medicine in Drug Discovery 6 (2020) 100024
[79] Zhao PF, Yin WM, Wu AH, Tang YS, Wang JY, Pan ZZ, et al. Dual-targeting to Can- [97] Chen W, Shi K, Chu B, Wei X, Qian Z. Mitochondrial surface engineering for multidrug
cer cells and M2 macrophages via biomimetic delivery of Mannosylated albumin resistance reversal. Nano Lett 2019;19:2905–13.
nanoparticles for drug-resistant Cancer therapy. Adv Funct Mater 2017;27: [98] Fang L, Qin X, Zhao J, Gou S. Construction of dual stimuli-responsive platinum(IV) hy-
1700403. brids with NQO1 targeting ability and overcoming Cisplatin resistance. Inorg Chem
[80] Xu F, Zhong H, Chang Y, Li D, Jin H, Zhang M, et al. Targeting death receptors for 2019;58:2191–200.
drug-resistant cancer therapy: Codelivery of pTRAIL and monensin using dual- [99] Xing Y, Ding T, Wang Z, Wang L, Guan H, Tang J, et al. Temporally controlled Photothermal/
targeting and stimuli-responsive self-assembling nanocomposites. Biomaterials 2018; photodynamic and combined therapy for overcoming multidrug resistance of Cancer by
158:56–73. Polydopamine Nanoclustered micelles. ACS Appl Mater Interfaces 2019;11:13945–53.
[81] Wang Z, He Q, Zhao W, Luo J, Gao W. Tumor-homing, pH- and ultrasound-responsive [100] Sangtani A, Petryayeva E, Susumu K, Oh E, Huston AL, Lasarte-Aragones G, et al. Nano-
polypeptide-doxorubicin nanoconjugates overcome doxorubicin resistance in cancer particle-peptide-drug bioconjugates for unassisted defeat of multidrug resistance in a
therapy. J Control Release 2017;264:66–75. model Cancer cell line. Bioconjug Chem 2019;30:525–30.
[82] Tang Y, Liang J, Wu A, Chen Y, Zhao P, Lin T, et al. Co-delivery of Trichosanthin and [101] Gao Y, Jia L, Wang Q, Hu H, Zhao X, Chen D, et al. pH/redox dual-responsive Polyplex
Albendazole by Nano-self-assembly for overcoming tumor multidrug-resistance and with effective Endosomal escape for Codelivery of siRNA and doxorubicin against
metastasis. ACS Appl Mater Interfaces 2017;9:26648–64. drug-resistant Cancer cells. ACS Appl Mater Interfaces 2019;11:16296–310.
[83] Suda K, Onozato R, Yatabe Y, Mitsudomi T. EGFR T790M mutation: a double role in [102] Qin Y, Zhang Z, Huang C, Fan F, Liu L, Lu L, et al. Folate-targeted redox-responsive
lung cancer cell survival? J Thorac Oncol 2009;4:1–4. Polymersomes loaded with chemotherapeutic drugs and Tariquidar to overcome
[84] Peng H, Chen B, Huang W, Tang Y, Jiang Y, Zhang W, et al. Reprogramming tumor-as- drug resistance. J Biomed Nanotechnol 2018;14:1705–18.
sociated macrophages to reverse EGFR(T790M) resistance by dual-targeting [103] Tong M, Gao M, Xu Y, Fu L, Li Y, Bao X, et al. SHR-A1403, a novel c-met antibody-drug
Codelivery of Gefitinib/Vorinostat. Nano Lett 2017;17:7684–90. conjugate, overcomes AZD9291 resistance in NSCLC cells overexpressing c-met. Can-
[85] Yin W, Yu X, Kang X, Zhao Y, Zhao P, Jin H, et al. Remodeling tumor-associated mac- cer Sci 2019;110:3584–94.
rophages and neovascularization overcomes EGFR(T790M) -associated drug resistance [104] Raoof S, Mulford IJ, Frisco-Cabanos H, Nangia V, Timonina D, Labrot E, et al. Targeting
by PD-L1 Nanobody-mediated Codelivery. Small 2018;14:e1802372. FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer.
[86] Teng MWL, Galon J, Fridman W-H, Smyth MJ. From mice to humans: developments in Oncogene 2019;38:6399–413.
cancer immunoediting. J Clin Investig 2015;125:3338–46. [105] Zhen S, Yi X, Zhao Z, Lou X, Xia F, Tang BZ. Drug delivery micelles with efficient near-
[87] Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from infrared photosensitizer for combined image-guided photodynamic therapy and che-
immunosurveillance to tumor escape. Nat Immunol 2002;3:991–8. motherapy of drug-resistant cancer. Biomaterials 2019;218:119330.
[88] Ribas A. Adaptive immune resistance: how Cancer protects from immune attack. Can- [106] Huang W, Zhao H, Wan J, Zhou Y, Xu Q, Zhao Y, et al. pH- and photothermal-driven
cer Discov 2015;5:915–9. multistage delivery nanoplatform for overcoming cancer drug resistance. Theranostics
[89] Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary. Adaptive, and Acquired Resis- 2019;9:3825–39.
tance to Cancer Immunotherapy, Cell 2017;168:707–23. [107] Zhou M, Li L, Li L, Lin X, Wang F, Li Q, et al. Overcoming chemotherapy resistance via
[90] Draghi A, Chamberlain CA, Furness A, Donia M. Acquired resistance to cancer immu- simultaneous drug-efflux circumvention and mitochondrial targeting. Acta Pharm Sin
notherapy. Semin Immunopathol 2019;41:31–40. B 2019;9:615–25.
[91] Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with com- [108] Hu Y, Zhang HR, Dong L, Xu MR, Zhang L, Ding WP, et al. Enhancing tumor chemo-
bination immunotherapies. Nat Rev Drug Discov 2019;18:197–218. therapy and overcoming drug resistance through autophagy-mediated intracellular
[92] Abdullah LN, Chow EK. Mechanisms of chemoresistance in cancer stem cells. Clin dissolution of zinc oxide nanoparticles. Nanoscale 2019;11:11789–807.
Transl Med 2013;2:3. [109] Qie S, Yoshida A, Parnham S, Oleinik N, Beeson GC, Beeson CC, et al. Targeting
[93] Lin J, Xia L, Liang J, Han Y, Wang H, Oyang L, et al. The roles of glucose metabolic glutamine-addiction and overcoming CDK4/6 inhibitor resistance in human esopha-
reprogramming in chemo- and radio-resistance. J Exp Clin Cancer Res 2019;38: geal squamous cell carcinoma. Nat Commun 2019;10:1296.
218. [110] He Y, Li X, Ma J, Ni G, Yang G, Zhou S. Programmable Codelivery of doxorubicin and
[94] Kelly B, O'Neill LA. Metabolic reprogramming in macrophages and dendritic cells in in- Apatinib using an implantable hierarchical-structured Fiber device for overcoming
nate immunity. Cell Res 2015;25:771–84. Cancer multidrug resistance. Small 2019;15:e1804397.
[95] Wang H, Tang Y, Fang Y, Zhang M, Wang H, He Z, et al. Reprogramming tumor im- [111] Chen M, Song F, Liu Y, Tian J, Liu C, Li R, et al. A dual pH-sensitive liposomal system
mune microenvironment (TIME) and metabolism via biomimetic targeting Codelivery with charge-reversal and NO generation for overcoming multidrug resistance in can-
of Shikonin/JQ1. Nano Lett 2019;19:2935–44. cer. Nanoscale 2019;11:3814–26.
[96] Li R, Xie Y. Nanodrug delivery systems for targeting the endogenous tumor microenvi- [112] Zhang L, Su H, Liu Y, Pang N, Li J, Qi XR. Enhancing solid tumor therapy with sequen-
ronment and simultaneously overcoming multidrug resistance properties. J Control tial delivery of dexamethasone and docetaxel engineered in a single carrier to over-
Release 2017;251:49–67. come stromal resistance to drug delivery. J Control Release 2019;294:1–16.
13