Nanomedicines for Overcoming Cancer Drug Resistance: Comparison
Please note this is a comparison between Version 2 by Ting Ting Hu and Version 4 by Jessie Wu.

Clinically, cancer drug resistance to chemotherapy, targeted therapy or immunotherapy remains the main impediment towards curative cancer therapy, which leads directly to treatment failure along with extended hospital stays, increased medical costs and high mortality. Therefore, increasing attention has been paid to nanotechnology-based delivery systems for overcoming drug resistance in cancer. In this respect, novel tumor-targeting nanomedicines offer fairly effective therapeutic strategies for surmounting the various limitations of chemotherapy, targeted therapy and immunotherapy, enabling more precise cancer treatment, more convenient monitoring of treatment agents, as well as surmounting cancer drug resistance, including multidrug resistance (MDR). Nanotechnology-based delivery systems, including liposomes, polymer micelles, nanoparticles (NPs), and DNA nanostructures, enable a large number of properly designed therapeutic nanomedicines. Nanomedicines have paved the way for effective treatment of cancer by rationally designing strategies such as passive targeted drug delivery, active targeted drug delivery, co-delivery of combinatorial agents and multimodal combination therapy, and have broad prospects in overcoming drug resistance. It is believed that nanomedicines will be an attractive strategy for reversing or overcoming cancer drug resistance.

  • nanomedicine
  • chemotherapy
  • drug

1. Mechanisms in Drug Resistance of Chemotherapy

Nowadays, chemotherapy is still the most widely used strategy for treating cancer; however, the biggest obstacle to this traditional strategy is the development of cancer drug resistance [1][2][3]. The mechanisms of drug resistance to chemotherapy are extremely complex [3]. Generally, the emergence of chemoresistance may be classified by the following pathways: (1) increased drug efflux by ATP-dependent pumps mediated by transmembrane transporters of the ATP-binding cassette (ABC) superfamily [4][5][6]; (2) reduced drug uptake mediated by altering specific cellular targets [7][8][9]; (3) inactivation of apoptotic pathways mediated by high expression of the Bcl-2 antiapoptotic family such as Bcl-2, Mcl-1 and Bcl-XL, which are mainly responsible for the reason why cancer cells can resist apoptosis [10][11][12]; (4) enhanced DNA repair ability that can contribute to the resistance of cancer by promoting genomic instability and mutation [13][14][15]; (5) alterations in specific drug targets [16][17]; (6) increased drug detoxification mediated by metabolism or biotransformation [18][19]. All in all, these resistance mechanisms can allow cancer cells to survive by easily changing different pathways, and ultimately resulting in chemotherapeutic failure.

2. Nanomedicines to Overcome Chemotherapy Resistance

Considering that chemotherapy resistance-related drug efflux proteins mainly reside in the nuclear membranes and blood, but not in the mitochondria [20][21], delivering chemotherapy agents into the mitochondria is an emerging strategy to surmount drug resistance to chemotherapy [22][23][24][25][26][27][28]. Yu et al. [29] constructed a weak acid-activated, charge-reversible, triphenylphosphonium (TPP)-based, “shell–core” nanosystem (DOX-PLGA/CPT/PD) for sequential facilitation of tumor accumulation, cellular uptake, mitochondria targeting, intracellular localization and surmounting drug resistance of MCF-7/ADR breast cancer. Firstly, positively charged mitochondrial-targeting lipid-polymer hybrid nanoparticles (PLGA/CPT) were prepared from PLGA and C18-PEG2000-TPP (CPT) [30]. Then, DOX was loaded into PLGA/CPT nanoparticles to obtain DOX-PLGA/CPT. Lastly, positively charged PEI-DMMA (PD) shell was wrapped on the surface of positively charged DOX-PLGA/CPT to obtain negatively charged DOX-PLGA/CPT/PD with a diameter of ~150 nm. When DOX-PLGA/CPT/PD was treated at pH 6.5, the hydrolysis of amide in PD occurred, facilitating the elimination of electrostatic interaction between PLGA/CPT and PEI, ultimately resulting in the deshielding of PD to reveal DOX-PLGA/CPT and transformation of the charge from −24 to +19.2 mV. Then, they studied the pharmacokinetics of DOX-PLGA/CPT/PD, and, the results showed that DOX-PLGA/CPT showed significantly slower clearance with a half-life time 15.84 h. After incubation with MCF-7/ADR cells at pH 6.5, DOX-PLGA/CPT/PD showed effective lysosome escape, excellent mitochondrial-targeting capacity and superior cytotoxicity for overcoming DOX resistance by up-regulating the apoptosis-related proteins as well as down-regulating the antiapoptotic protein Bcl-2. Encouraged by the in vitro antitumor effect of DOX-PLGA/CPT/PD, Yu et al. evaluated the in vivo effect in MCF-7/ADR cell-bearing mice. The results show that DOX-PLGA/CPT/PD showed the best inhibitory effect on tumor growth and exhibited the best treatment effect, with a tumor inhibition rate (TIR) of 84.9% with no obvious side effects.
Studies show that the exposure of tumor cells to chemotherapy drugs can result in hypoxia-inducible factor-1 (HIF-1) activation and stabilization [31][32], where HIF-1 plays an important part in drug resistance by regulating multidrug resistance protein (MRP), P-glycoprotein (P-gp), Bcl-2, etc. [33][34][35]. Moreover, HIF-1 can up-regulate the level of glutathione, which can bind with heavy metal ions, including cisplatin [36][37]. Therefore, inhibiting HIF-1 pathways during chemotherapy might be a promising method to circumvent chemo-resistance [38][39][40][41][42]. Acriflavine (ACF), a potent HIF-1 inhibitor, has been proven to bind to HIF-1α and thereby impede HIF-1α/β dimerization [43][44], which can be a useful strategy for sensitization of chemotherapy. In this regard, Zhang et al. [45] developed a new type of microporous silica-based co-delivery system (PMONA) to reverse the acquired resistance to cisplatin. Firstly, cisplatin was loaded into the polymeric mPEG-silane functionalized mesoporous silica nanoparticles inner core by reverse microemulsion method, where polymeric mPEG-silane was applied to maintain stability during blood circulation. To achieve tumor-specific glutathione (GSH)-triggered drug release, tetrasulfide bond-bridged organosilica was integrated to obtain the nanoparticles. Finally, ACF was loaded into the inner area of mircopores by electrostatic interactions to obtain ACF-loaded nanoparticles with a diameter of ~45 nm. After internalization by cancer cells, the outer organosilica shell of PMONA could be degraded by intracellular GSH, resulting in nanoparticle disassembly, drug release and synergistic regulation of multiple cancer-related signaling pathways. As shown in an in vitro release experiment, cisplatin and ACF had faster and higher cumulative release rates in a medium containing 10 mM GSH than in a medium containing 10 μM GSH, which confirmed that the tetrasulfide bond in organosilica enabled GSH-responsive disassembly and drug release. After incubation with A459 cells, PMONA exhibited stronger cell cytotoxicity, induced more apoptosis than the single drug-loaded nanoparticles by suppressing HIF-1-related proteins and decreased the level of intracellular GSH. Inspired by the result that ACF strengthens the curative effect of cisplatin in vitro, Zhang et al. assessed the in vivo antitumor effect in A459 cell-bearing mice. The results indicated that PMONA showed the best inhibitory effect on tumor growth and exhibited the best therapeutic effect with limited side effects. Additionally, the immunohistochemical experiment showed that PMONA enhanced cell death and apoptosis in tumor tissues mainly by down-regulating the levels of P-gp, MRP2, HIF-1-activated glutamate-cysteine ligase modifier subunit (GCLM), vascular endothelial growth factor (VEGF) and cystine transporter (xCT). Taken together, these results confirmed that ACF could combat cisplatin-acquired resistance by inhibiting HIF-1 function.
Hyperthermia, a non-invasive treatment strategy, has shown a competitive advantage in reversing drug resistance in cancer by suppressing the expression of drug efflux transporters [46][47][48][49][50][51]. Therefore, hyperthermia combined with chemotherapy is a hopeful treatment strategy for overcoming chemotherapeutic resistance [52][53][54][55][56][57]. Huang et al. [58] constructed smart, thermoresponsive, pH low insertion peptide (pHLIP)-modified gold nanocages (DOX@pPGNCs) to realize synergistic thermo-chemotherapy and overcome chemotherapeutic resistance. Firstly, thermoresponsive poly (di (ethylene glycol) methyl ether methacrylate-co-oligo (ethylene glycol) methyl ether methacrylate) (PMEO2MA-OEGMA) polymer was anchored to gold nanocages to PMEO2MA-OEGMA-modified gold nanocages, where PMEO2MA-OEGMA served as a temperature-sensitive gate guard at a lower critical solution temperature of ca. 41.6 °C. In other words, the PMEO2MA-OEGMA chains extended under 41.6 °C, sealing the pore of gold nanocages to prevent the leakage of drug into the blood; however, once the temperature increased up to 41.6 °C due to the NIR-induced photothermal effects, its chains shrunk, leading to opening of the pores of gold nanocages and fast DOX release. Then, pHLIP was used to decorate PMEO2MA-OEGMA-modified gold nanocages to obtain pPGNCs, where pHLIP was a good candidate to enhance cancer cell internalization by conformational transition at the weakly acidic tumor microenvironment. Lastly, DOX was loaded into pPGNCs to obtain DOX@pPGNCs with a diameter of ~160 nm and a zeta potential of approximately −20 mV. PMEO2MA-OEGMA was thermosensitive with a lower critical solution temperature of ca. 41.6 °C. In vitro release experiments indicated that the cumulative release of DOX increased from 3.7 to 20.1% after 5 min of NIR irradiation. More importantly, the rapid release of DOX was consistent under NIR irradiation in another cycle, indicating that PMEO2MA-OEGMA was a very responsible gatekeeper to precisely control NIR-triggered DOX release from DOX@pPGNCs. Cytotoxicity experiments showed that the antiproliferation ability against MCF-7/ADR cells was strongest in the DOX@pPGNCs and NIR irradiation group at pH 6.5, suggesting that pHLIP could enhance cellular uptake of DOX@pPGNCs under a weak acid tumor microenvironment, and, upon NIR irradiation, DOX@pPGNCs could efficiently achieve synergistic thermo-chemotherapy to overcome cancer resistance. In vivo biodistribution experiments showed that DOX accumulation in tumor site of tumor-bearing mice treated with DOX@ pPGNCs and NIR irradiation was highest, confirming that NIR irradiation-triggered photothermal effects of gold nanocages could further strengthen DOX accumulation. Inspired by the above experimental results, Huang et al. further assessed the in vivo treatment effect in MCF-7/ADR cell-bearing mice. The results indicated that DOX@pPGNCs achieved the strongest antitumor efficacy with a TIR of 97.3%, indicating the highly effective synergistic thermo-chemotherapy in MCF-7/ADR cell-bearing mice.
Studies have shown that tumor cells can develop drug resistance by enhancing DNA repair [59][60][61][62][63][64], suggesting that drug resistance owing to DNA repair can be overcome by inhibiting the function of related proteins [65][66][67][68]. Recently, Wang et al. [69] constructed a smart delivery system for overcoming cisplatin-related “cascade drug resistance” (CDR) by mild hyperthermia (43 °C) triggered by NIR. Firstly, hydrophobic photothermal-conjugated polymer and biodegradable amphiphilic polymer were mixed to form F-nanoparticles (F-NPs) with photothermal performance. Secondly, biodegradable amphiphilic polymer and C16-CisPt-Suc (a Pt (IV) prodrug) were mixed to form Pt-nanoparticles (Pt-NPs). Lastly, Pt-NPs and F-NPs were mixed to obtain the mixed nanoparticles (F-Pt-NPs). On the basis of DLS data, the average particle size of F-NPs was 91.0 ± 2.6 nm, while that of the Pt-NPs was 105.1 ± 1.6 nm. In vitro experiments showed that, under the treatment of NIR, mild hyperthermia could efficiently facilitate cellular uptake of drug-resistant A549DDP cells, resulting in enhanced cytotoxicity and surmounting CDR of cisplatin by the consumption of GSH and the reduction of Pt (IV) to Pt (II). More importantly, mild hyperthermia could accelerate the binding of Pt to DNA and promote the formation of irreparable crosslinking of Pt-DNA strands, resulting in the destruction of DNA repair. In vivo experiments showed that, under mild hyperthermia conditions, F-Pt-NPs exhibited the best antitumor effect with a TIR of 94% with few side effects, further indicating that NIR-triggered mild hyperthermia could reverse CDR.
In recent years, substantial evidence has confirmed that drug resistance is closely related to the CSC phenotype [70][71]. One proven mechanism of multidrug resistance (MDR) in CSC is the increased expression of ABC transporters [72]. In addition, the CSC phenotype shows increased drug resistance to chemotherapy by modulating many other stem characteristics, including enhanced DNA damage repair capacity and up-regulation of antiapoptotic proteins [73][74]. Therefore, eradication of CSCs is an effective strategy to surmounting cancer drug resistance. Shen et al. [75] constructed an alltrans-retinoic acid (ATRA) and camptothecin (CPT) co-loaded nanoplatform (ATRA/CPT-NPs) to surmount chemotherapeutic resistance of both CSCs and bulk tumor cells. Firstly, ROS-responsive nitroimidazole-modified hyaluronic acid-oxalate-CPT conjugate (n-HA-oxa-CPT) was synthesized. Then, n-HA-oxa-CPT assembled into nanoparticles and physically encapsulated ATRA to obtain ATRA/CPT-NPs with a diameter of ~150 nm. Based on the difference levels of ROS between bulk tumor cells and CSCs, ATRA/CPT-NPS could sequentially release ATRA and CPT during the differentiation of CSCs. After uptake by hypoxia CSCs, ATRA was firstly released, which induced CSC differentiation into reduced stemness and chemoresistance, along with increased ROS level. Then, the increased ROS in differentiated CSCs triggered CPT release for enhanced cytotoxicity towards the differentiated cells with decreased drug resistance. On the other hand, after uptake by bulk tumor cells with hypoxia and high ROS, ATRA/CPT-NPS could simultaneously release ATRA and CPT, resulting powerful synergistic anticancer effects. In their study, ATRA/CPT-NPs showed the strongest inhibition efficacy on the orthotopic BCSC-enriched tumor mouse models, suggesting that the differential drug release realized by ATRA/CPT-NPs was very important to strengthen the synergistic efficacy of ATRA-triggered CSC differentiation and CPT-triggered cytotoxic activity for the treatment of poorly differentiated and highly chemo-resistant heterogeneous tumors.
Additionally, a summary of nanomedicines studied for overcoming chemotherapeutic resistance in recent years is displayed in Table 1.
Table 1.
 Recent advances in nanomedicines for overcoming chemotherapeutic resistance.
Nanoformulation Name Particle Size Payload Reversal Mechanism of Drug Resistance Cell Line Tumor Model Reference
Polymeric micelles ACP-Dox and Apa micelles 104 ± 2 nm DOX and apatinib Inhibit P-gp activity MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [76]
HA-PLGA (PTX and FAK siRNA)-NPs 232.9 ± 6.9 nm PTX and FAK siRNA siRNA-mediated silencing of FAK HeyA8-MDR and SKOV3-TR cells Drug-resistant, patient-derived xenograft (PDX) model [77]
ACP-R837 and PPP-DOX ~110 nm R837 and DOX Synergistic chemo-immunotherapy 4T1 cells 4T1 tumor-bearing mice [78]
NC-DOX ~122 nm DOX and IR780 Combined chemotherapy/PTT/PDT MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [79]
Polymeric nanoparticles Dox-Cur-NDs 55.1 ± 3.0 nm DOX and CUR Down-regulate the expression of P-gp A2780 ADR cells A2780 ADR tumor-bearing mice [80]
[FeFe]TPP/GEM/FCS NPs 176.0 ± 17.2 nm Gemcitabine and [FeFe]TPP Reduce the of function P-gp efflux pump T24 cells T24 tumor-bearing mice [81]
IGU-PLGA-NPs 199.6 nm Iguratimod Facilitate BBB penetration and inhibit GSCs proliferation and stemness U87 and U251TMZ-R cells U87 tumor-bearing mice [82]
Liposomes rTLM-PEG, PTX liposomes / PTX and trichosanthin Reverse caspase 9 phosphorylation and induce caspase 3-dependent apoptosis A549/T cells A549/T tumor-bearing mice [83]
PTX/NO/DMA-L 146.3 ± 0.82 nm PTX and DETA NONOate NO-mediated down-regulation of P-gp A549/T cells A549/T tumor-bearing mice [84]
CBZ liposomes 108.53 ± 1.5 nm CBZ G2/M phase arrest MCF-7 and MDA-MB-231 cells Female SD rats [85]
Lip (Ap-Dox) 128.6 nm Ap-Dox complex Bypass the P-gp-mediated drug efflux MCF-7/ADR cells MCF-7/ADR tumor-bearing nude mice [86]
(DEX and DTX)-Lip 74.02 ± 0.41 nm DTX and dexamethasone Overcome stroma obstacles Multidrug-resistant KBv cells and 4 T1 cells Multidrug-resistant KBv and metastatic 4 T1 tumor models [87]
FPL-DOX/IM 159 ± 6 nm DOX and imatinib Inhibit ABC transporter function MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [88]
PpIX/Dox liposomes 55.9 ± 20.9 nm DOX and PpIX Disrupt the structure of P-gp MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [89]
Nanogels LNGs-PTX-siRNA ~100 nm PTX and MDR1 siRNA Knockdown MDR1 DROV cells DROV tumor-bearing mice [90]
CDDP/DOX-NGs ~100 nm CDDP and DOX Combination chemotherapy MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [91]
HA/Cis/Dox 45 ± 9.9 nm DOX GSH-induced DOX release A2780cis cells / [92]
SiPT75 75.5 ± 19.8 nm TPPS Elude the drug efflux pumps and retards exocytosis of cells A549/DDP cells A549/DDP tumor-bearing mice [93]
Inorganic nanoparticles H-MSNs-DOX/siRNA nanoparticles ~100 nm P-gp siRNA and DOX siRNA-mediated silencing of P-gp MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [94]
Pt-AuNS ~85 nm Pt GSH depletion and GPX4 inactivation MCF-7/ADR cells MCF-7/ADR tumor-bearing mice [95]
FA-GT-MSNs@TPZ ~60 nm TPZ Synergistic radio-chemo-photothermal therapy Hypoxic SMMC-7721 cells SMMC-7721 tumor-bearing mice [96]
Hybrid nanoparticles SCA4PNPBTZ ~150 nm BTZ and CA4P Inhibit the overexpression of BCRP/ABCG2 A549 cells Human A549 pulmonary adenocarcinoma xenograft model and PDX model of colon cancer [97]
cNPs 286 ± 79 nm Afatinib, rapamycin and docetaxel Synergistic treatment HER2-positive breast cancer cells, EGFR-positive NSCLC cells and SKBR-3/AR cell lines HER2-positive breast cancer mouse model [98]
4T1-HANG-GNR-DC 103.1 ± 7.6 nm CDDP and DOX Synergistic chemo-photothermal therapy 4T1 cells 4T1 tumor-bearing mice [99]
IR780/DTX-PCEC@RBC ~150 nm IR780 and DTX Combination therapy MCF-7 cells MCF-7 tumor-bearing mice [100]
cNC@PDA-PEG 170.5 ± 1.4 nm Paclitaxel/lapatinib Combination therapy MCF-7/ADR cells / [101]
miR497/TP-HENPs 125 ± 6 nm miR497 and triptolide Synergically suppress mTOR signaling pathway SKOV3-CDDP cells SKOV3-CDDP tumor-bearing mice [102]
“/”: The original research article did not mention it.
 

References

  1. Bar-Zeev, M.; Livney, Y.D.; Assaraf, Y.G. Targeted nanomedicine for cancer therapeutics: Towards precision medicine overcoming drug resistance. Drug Resist. Updat. 2017, 31, 15–30.
  2. Lepeltier, E.; Rijo, P.; Rizzolio, F.; Popovtzer, R.; Petrikaite, V.; Assaraf, Y.G.; Passirani, C. Nanomedicine to target multidrug resistant tumors. Drug Resist. Updat. 2020, 52, 100704.
  3. Wei, X.; Song, M.; Li, W.; Huang, J.; Yang, G.; Wang, Y. Multifunctional nanoplatforms co-delivering combinatorial dual-drug for eliminating cancer multidrug resistance. Theranostics 2021, 11, 6334–6354.
  4. Chen, Z.S.; Tiwari, A.K. Multidrug resistance proteins (MRPs/ABCCs) in cancer chemotherapy and genetic diseases. FEBS J. 2011, 278, 3226–3245.
  5. Amawi, H.; Sim, H.M.; Tiwari, A.K.; Ambudkar, S.V.; Shukla, S. ABC transporter-mediated multidrug-resistant cancer. Adv. Exp. Med. Biol. 2019, 1141, 549–580.
  6. Muriithi, W.; Macharia, L.W.; Heming, C.P.; Echevarria, J.L.; Nyachieo, A.; Filho, P.N.; Neto, V.M. ABC transporters and the hallmarks of cancer: Roles in cancer aggressiveness beyond multidrug resistance. Cancer Biol. Med. 2020, 17, 253–269.
  7. Rothem, L.; Ifergan, I.; Kaufman, Y.; Priest, D.G.; Jansen, G.; Assaraf, Y.G. Resistance to multiple novel antifolates is mediated via defective drug transport resulting from clustered mutations in the reduced folate carrier gene in human leukaemia cell lines. Biochem. J. 2002, 367, 741–750.
  8. Bosson, G. Reduced folate carrier: Biochemistry and molecular biology of the normal and methotrexate-resistant cell. Br. J. Biomed. Sci. 2003, 60, 117–129.
  9. Kordus, S.L.; Baughn, A.D. Revitalizing antifolates through understanding mechanisms that govern susceptibility and resistance. Medchemcomm 2019, 10, 880–895.
  10. Zhang, Z.; Bai, L.; Hou, L.; Deng, H.; Luan, S.; Liu, D.; Huang, M.; Zhao, L. Trends in targeting Bcl-2 anti-apoptotic proteins for cancer treatment. Eur. J. Med. Chem. 2022, 232, 114184.
  11. Cory, S.; Roberts, A.W.; Colman, P.M.; Adams, J.M. Targeting BCL-2-like proteins to kill cancer cells. Trends Cancer 2016, 2, 443–460.
  12. Sarosiek, K.A.; Letai, A. Directly targeting the mitochondrial pathway of apoptosis for cancer therapy using BH3 mimetics-recent successes, current challenges and future promise. FEBS J. 2016, 283, 3523–3533.
  13. Li, L.Y.; Guan, Y.D.; Chen, X.S.; Yang, J.M.; Cheng, Y. DNA repair pathways in cancer therapy and resistance. Front. Pharmacol. 2021, 11, 629266.
  14. Alhmoud, J.F.; Woolley, J.F.; Al Moustafa, A.E.; Malki, M.I. DNA damage/repair management in cancers. Cancers 2020, 12, 1050.
  15. Goldstein, M.; Kastan, M.B. The DNA damage response: Implications for tumor responses to radiation and chemotherapy. Annu. Rev. Med. 2015, 66, 129–143.
  16. Dal Bo, M.; De Mattia, E.; Baboci, L.; Mezzalira, S.; Cecchin, E.; Assaraf, Y.G.; Toffoli, G. New insights into the pharmacological, immunological, and CAR-T-cell approaches in the treatment of hepatocellular carcinoma. Drug Resist. Updat. 2020, 51, 100702.
  17. Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726.
  18. Dallavalle, S.; Dobričić, V.; Lazzarato, L.; Gazzano, E.; Machuqueiro, M.; Pajeva, I.; Tsakovska, I.; Zidar, N.; Fruttero, R. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist. Updat. 2020, 50, 100682.
  19. Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160.
  20. Dartier, J.; Lemaitre, E.; Chourpa, I.; Goupille, C.; Servais, S.; Chevalier, S.; Mahéo, K.; Dumas, J.F. ATP-dependent activity and mitochondrial localization of drug efflux pumps in doxorubicin-resistant breast cancer cells. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1075–1084.
  21. Ruan, L.; Chen, J.; Du, C.; Lu, H.; Zhang, J.; Cai, X.; Dou, R.; Lin, W.; Chai, Z.; Nie, G.; et al. Mitochondrial temperature-responsive drug delivery reverses drug resistance in lung cancer. Bioact. Mater. 2021, 13, 191–199.
  22. Cheng, F.; Pan, Q.; Gao, W.; Pu, Y.; Luo, K.; He, B. Reversing chemotherapy resistance by a synergy between lysosomal pH-activated mitochondrial drug delivery and erlotinib-mediated drug efflux inhibition. ACS Appl. Mater. Interfaces 2021, 13, 29257–29268.
  23. Zhou, M.; Li, L.; Li, L.; Lin, X.; Wang, F.; Li, Q.; Huang, Y. Overcoming chemotherapy resistance via simultaneous drug-efflux circumvention and mitochondrial targeting. Acta Pharm. Sin. B 2019, 9, 615–625.
  24. Zhang, Y.; Zhang, C.; Chen, J.; Liu, L.; Hu, M.; Li, J.; Bi, H. Trackable mitochondria-targeting nanomicellar loaded with doxorubicin for overcoming drug resistance. ACS Appl. Mater. Interfaces 2017, 9, 25152–25163.
  25. Liang, L.; Peng, Y.; Qiu, L. Mitochondria-targeted vitamin E succinate delivery for reversal of multidrug resistance. J. Control. Release 2021, 337, 117–131.
  26. Dong, X.; Sun, Y.; Li, Y.; Ma, X.; Zhang, S.; Yuan, Y.; Kohn, J.; Liu, C.; Qian, J. Synergistic combination of bioactive hydroxyapatite nanoparticles and the chemotherapeutic doxorubicin to overcome tumor multidrug resistance. Small 2021, 17, e2007672.
  27. Gao, D.; Zhu, Q.; Ruan, J.; Sun, T.; Han, L. Polyplexes by polymerized dequalinium and bifunctional aptamer for mitochondrial targeting drug release to overcome drug resistance. ACS Appl. Bio. Mater. 2020, 3, 5182–5192.
  28. Liu, Y.; Zhang, X.; Zhou, M.; Nan, X.; Chen, X.; Zhang, X. Mitochondrial-targeting lonidamine-doxorubicin nanoparticles for synergistic chemotherapy to conquer drug resistance. ACS Appl. Mater. Interfaces 2017, 9, 43498–43507.
  29. Yu, H.; Li, J.M.; Deng, K.; Zhou, W.; Wang, C.X.; Wang, Q.; Li, K.H.; Zhao, H.Y.; Huang, S.W. Tumor acidity activated triphenylphosphonium-based mitochondrial targeting nanocarriers for overcoming drug resistance of cancer therapy. Theranostics 2019, 9, 7033–7050.
  30. Zhou, W.; Yu, H.; Zhang, L.J.; Wu, B.; Wang, C.X.; Wang, Q.; Deng, K.; Zhuo, R.X.; Huang, S.W. Redox-triggered activation of nanocarriers for mitochondria-targeting cancer chemotherapy. Nanoscale 2017, 9, 17044–17053.
  31. Xiang, L.; Wang, Y.; Lan, J.; Na, F.; Wu, S.; Gong, Y.; Du, H.; Shao, B.; Xie, G. HIF-1-dependent heme synthesis promotes gemcitabine resistance in human non-small cell lung cancers via enhanced ABCB6 expression. Cell. Mol. Life Sci. 2022, 79, 343.
  32. Samanta, D.; Gilkes, D.M.; Chaturvedi, P.; Xiang, L.; Semenza, G.L. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, E5429–E5438.
  33. Karakashev, S.V.; Reginato, M.J. Progress toward overcoming hypoxia-induced resistance to solid tumor therapy. Cancer Manag. Res. 2015, 7, 253–264.
  34. Ghattass, K.; Assah, R.; El-Sabban, M.; Gali-Muhtasib, H. Targeting hypoxia for sensitization of tumors to radio- and chemotherapy. Curr. Cancer Drug Targets 2013, 13, 670–685.
  35. Albadari, N.; Deng, S.; Li, W. The transcriptional factors HIF-1 and HIF-2 and their novel inhibitors in cancer therapy. Expert Opin. Drug Discov. 2019, 14, 667–682.
  36. Lu, H.; Samanta, D.; Xiang, L.; Zhang, H.; Hu, H.; Chen, I.; Bullen, J.W.; Semenza, G.L. Chemotherapy triggers HIF-1-dependent glutathione synthesis and copper chelation that induces the breast cancer stem cell phenotype. Proc. Natl. Acad. Sci. USA 2015, 112, E4600–E4609.
  37. Luo, K.; Guo, W.; Yu, Y.; Xu, S.; Zhou, M.; Xiang, K.; Niu, K.; Zhu, X.; Zhu, G.; An, Z.; et al. Reduction-sensitive platinum (IV)-prodrug nano-sensitizer with an ultra-high drug loading for efficient chemo-radiotherapy of Pt-resistant cervical cancer in vivo. J. Control. Release 2020, 326, 25–37.
  38. Li, J.; Xi, W.; Li, X.; Sun, H.; Li, Y. Advances in inhibition of protein-protein interactions targeting hypoxia-inducible factor-1 for cancer therapy. Bioorg. Med. Chem. 2019, 27, 1145–1158.
  39. Wang, X.; Du, Z.W.; Xu, T.M.; Wang, X.J.; Li, W.; Gao, J.L.; Li, J.; Zhu, H. HIF-1α is a rational target for future ovarian cancer therapies. Front. Oncol. 2021, 11, 785111.
  40. Shirai, Y.; Chow, C.C.T.; Kambe, G.; Suwa, T.; Kobayashi, M.; Takahashi, I.; Harada, H.; Nam, J.M. An overview of the recent development of anticancer agents targeting the HIF-1 transcription factor. Cancers 2021, 13, 2813.
  41. Tang, W.; Zhao, G. Small molecules targeting HIF-1α pathway for cancer therapy in recent years. Bioorg. Med. Chem. 2020, 28, 115235.
  42. Ma, Z.; Xiang, X.; Li, S.; Xie, P.; Gong, Q.; Goh, B.C.; Wang, L. Targeting hypoxia-inducible factor-1, for cancer treatment: Recent advances in developing small-molecule inhibitors from natural compounds. Semin. Cancer Biol. 2022, 80, 379–390.
  43. Montigaud, Y.; Ucakar, B.; Krishnamachary, B.; Bhujwalla, Z.M.; Feron, O.; Préat, V.; Danhier, F.; Gallez, B.; Danhier, P. Optimized acriflavine-loaded lipid nanocapsules as a safe and effective delivery system to treat breast cancer. Int. J. Pharm. 2018, 551, 322–328.
  44. Weijer, R.; Broekgaarden, M.; Krekorian, M.; Alles, L.K.; van Wijk, A.C.; Mackaaij, C.; Verheij, J.; van der Wal, A.C.; van Gulik, T.M.; Storm, G.; et al. Inhibition of hypoxia inducible factor 1 and topoisomerase with acriflavine sensitizes perihilar cholangiocarcinomas to photodynamic therapy. Oncotarget 2016, 7, 3341–3356.
  45. Zhang, X.; He, C.; Liu, X.; Chen, Y.; Zhao, P.; Chen, C.; Yan, R.; Li, M.; Fan, T.; Altine, B.; et al. One-pot synthesis of a microporous organosilica-coated cisplatin nanoplatform for HIF-1-targeted combination cancer therapy. Theranostics 2020, 10, 2918–2929.
  46. Liu, Y.; Bao, Q.; Chen, Z.; Yao, L.; Ci, Z.; Wei, X.; Wu, Y.; Zhu, J.; Sun, K.; Zhou, G.; et al. Circumventing drug resistance pathways with a nanoparticle-based photodynamic method. Nano Lett. 2021, 21, 9115–9123.
  47. Li, Y.; Deng, Y.; Tian, X.; Ke, H.; Guo, M.; Zhu, A.; Yang, T.; Guo, Z.; Ge, Z.; Yang, X.; et al. Multipronged design of light-triggered nanoparticles to overcome cisplatin resistance for efficient ablation of resistant tumor. ACS Nano 2015, 9, 9626–9637.
  48. Yang, G.G.; Pan, Z.Y.; Zhang, D.Y.; Cao, Q.; Ji, L.N.; Mao, Z.W. Precisely assembled nanoparticles against cisplatin resistance via cancer-specific targeting of mitochondria and imaging-guided chemo-photothermal therapy. ACS Appl. Mater. Interfaces 2020, 12, 43444–43455.
  49. Wang, T.; Wang, D.; Yu, H.; Wang, M.; Liu, J.; Feng, B.; Zhou, F.; Yin, Q.; Zhang, Z.; Huang, Y.; et al. Intracellularly acid-switchable multifunctional micelles for combinational photo/chemotherapy of the drug-resistant tumor. ACS Nano 2016, 10, 3496–3508.
  50. Souslova, T.; Averill-Bates, D.A. Multidrug-resistant hela cells overexpressing MRP1 exhibit sensitivity to cell killing by hyperthermia: Interactions with etoposide. Int. J Radiat. Oncol. Biol. Phys. 2004, 60, 1538–1551.
  51. Stein, U.; Jürchott, K.; Walther, W.; Bergmann, S.; Schlag, P.M.; Royer, H.D. Hyperthermia-induced nuclear translocation of transcription factor YB-1 leads to enhanced expression of multidrug resistance-related ABC transporters. J. Biol. Chem. 2001, 276, 28562–28569.
  52. Nair, J.B.; Joseph, M.M.; Arya, J.S.; Sreedevi, P.; Sujai, P.T.; Maiti, K.K. Elucidating a thermoresponsive multimodal photo-chemotherapeutic nanodelivery vehicle to overcome the barriers of doxorubicin therapy. ACS Appl. Mater. Interfaces 2020, 12, 43365–43379.
  53. Jiang, D.; Xu, M.; Pei, Y.; Huang, Y.; Chen, Y.; Ma, F.; Lu, H.; Chen, J. Core-matched nanoassemblies for targeted co-delivery of chemotherapy and photosensitizer to treat drug-resistant cancer. Acta Biomater. 2019, 88, 406–421.
  54. Jiang, D.; Gao, X.; Kang, T.; Feng, X.; Yao, J.; Yang, M.; Jing, Y.; Zhu, Q.; Feng, J.; Chen, J. Actively targeting D-α-tocopheryl polyethylene glycol 1000 succinate-poly(lactic acid) nanoparticles as vesicles for chemo-photodynamic combination therapy of doxorubicin-resistant breast cancer. Nanoscale 2016, 8, 3100–3118.
  55. Li, Z.; Wang, H.; Chen, Y.; Wang, Y.; Li, H.; Han, H.; Chen, T.; Jin, Q.; Ji, J. pH- and NIR light-responsive polymeric prodrug micelles for hyperthermia-assisted site-specific chemotherapy to reverse drug resistance in cancer treatment. Small 2016, 12, 2731–2740.
  56. Gaio, E.; Conte, C.; Esposito, D.; Miotto, G.; Quaglia, F.; Moret, F.; Reddi, E. Co-delivery of docetaxel and disulfonate tetraphenyl chlorin in one nanoparticle produces strong synergism between chemo- and photodynamic therapy in drug-sensitive and -resistant cancer cells. Mol. Pharm. 2018, 15, 4599–4611.
  57. Shi, C.; Huang, H.; Zhou, X.; Zhang, Z.; Ma, H.; Yao, Q.; Shao, K.; Sun, W.; Du, J.; Fan, J.; et al. Reversing multidrug resistance by inducing mitochondrial dysfunction for enhanced chemo-photodynamic therapy in tumor. ACS Appl. Mater. Interfaces 2021, 13, 45259–45268.
  58. Huang, W.; Zhao, H.; Wan, J.; Zhou, Y.; Xu, Q.; Zhao, Y.; Yang, X.; Gan, L. pH- and photothermal-driven multistage delivery nanoplatform for overcoming cancer drug resistance. Theranostics 2019, 9, 3825–3839.
  59. Curtin, N.J. DNA repair dysregulation from cancer driver to therapeutic target. Nat. Rev. Cancer 2012, 12, 801–817.
  60. Yang, C.; Zang, W.; Tang, Z.; Ji, Y.; Xu, R.; Yang, Y.; Luo, A.; Hu, B.; Zhang, Z.; Liu, Z.; et al. A20/TNFAIP3 regulates the DNA damage response and mediates tumor cell resistance to DNA-damaging therapy. Cancer Res. 2018, 78, 1069–1082.
  61. Stover, E.H.; Konstantinopoulos, P.A.; Matulonis, U.A.; Swisher, E.M. Biomarkers of response and resistance to DNA repair targeted therapies. Clin. Cancer Res. 2016, 22, 5651–5660.
  62. Sadoughi, F.; Mirsafaei, L.; Dana, P.M.; Hallajzadeh, J.; Asemi, Z.; Mansournia, M.A.; Montazer, M.; Hosseinpour, M.; Yousefi, B. The role of DNA damage response in chemo- and radio-resistance of cancer cells: Can DDR inhibitors sole the problem? DNA Repair 2021, 101, 103074.
  63. Bouwman, P.; Jonkers, J. The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nat. Rev. Cancer 2012, 12, 587–598.
  64. Pilié, P.G.; Tang, C.; Mills, G.B.; Yap, T.A. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat. Rev. Clin. Oncol. 2019, 16, 81–104.
  65. Sharma, M.; Anand, P.; Padwad, Y.S.; Dogra, V.; Acharya, V. DNA damage response proteins synergistically affect the cancer prognosis and resistance. Free. Radic. Biol. Med. 2022, 178, 174–188.
  66. Li, L.; Kumar, A.K.; Hu, Z.; Guo, Z. Small molecule inhibitors targeting key proteins in the DNA damage response for cancer therapy. Curr. Med. Chem. 2021, 28, 963–985.
  67. Zhu, Y.; Hu, J.; Hu, Y.; Liu, W. Targeting DNA repair pathways: A novel approach to reduce cancer therapeutic resistance. Cancer Treat. Rev. 2009, 35, 590–596.
  68. Helleday, T.; Petermann, E.; Lundin, C.; Hodgson, B.; Sharma, R.A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer 2008, 8, 193–204.
  69. Wang, L.; Yu, Y.; Wei, D.; Zhang, L.; Zhang, X.; Zhang, G.; Ding, D.; Xiao, H.; Zhang, D. A systematic strategy of combinational blow for overcoming cascade drug resistance via NIR-light-triggered hyperthermia. Adv. Mater. 2021, 33, e2100599.
  70. Tang, Y.; Chen, Y.; Zhang, Z.; Tang, B.; Zhou, Z.; Chen, H. Nanoparticle-based RNAi therapeutics targeting cancer stem cells: Update and prospective. Pharmaceutics 2021, 13, 2116.
  71. Huang, T.; Song, X.; Xu, D.; Tiek, D.; Goenka, A.; Wu, B.; Sastry, N.; Hu, B.; Cheng, S.Y. Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics 2020, 10, 8721–8743.
  72. Dean, M.; Fojo, T.; Bates, S. Tumor stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284.
  73. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. 2018, 2018, 5416923.
  74. Garcia-Mayea, Y.; Mir, C.; Masson, F.; Paciucci, R.; LLeonart, M.E. Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin. Cancer Biol. 2020, 60, 166–180.
  75. Shen, S.; Xu, X.; Lin, S.; Zhang, Y.; Liu, H.; Zhang, C.; Mo, R. A nanotherapeutic strategy to overcome chemotherapeutic resistance of cancer stem-like cells. Nat. Nanotechnol. 2021, 16, 104–113.
  76. Wei, X.; Liu, L.; Guo, X.; Wang, Y.; Zhao, J.; Zhou, S. Light-activated ROS-responsive nanoplatform co-delivering apatinib and doxorubicin for enhanced chemo-photodynamic therapy of multidrug-resistant tumors. ACS Appl. Mater. Interfaces 2018, 10, 17672–17684.
  77. Byeon, Y.; Lee, J.W.; Choi, W.S.; Won, J.E.; Kim, G.H.; Kim, M.G.; Wi, T.I.; Lee, J.M.; Kang, T.H.; Jung, I.D.; et al. CD44-targeting PLGA nanoparticles incorporating paclitaxel and FAK siRNA overcome chemoresistance in epithelial ovarian cancer. Cancer Res. 2018, 78, 6247–6256.
  78. Wei, X.; Liu, L.; Li, X.; Wang, Y.; Guo, X.; Zhao, J.; Zhou, S. Selectively targeting tumor-associated macrophages and tumor cells with polymeric micelles for enhanced cancer chemo-immunotherapy. J. Control. Release 2019, 313, 42–53.
  79. Xing, Y.; Ding, T.; Wang, Z.; Wang, L.; Guan, H.; Tang, J.; Mo, D.; Zhang, J. Temporally controlled photothermal/photodynamic and combined therapy for overcoming multidrug resistance of cancer by polydopamine nanoclustered micelles. ACS Appl. Mater. Interfaces 2019, 11, 13945–13953.
  80. Baghbani, F.; Moztarzadeh, F. Bypassing multidrug resistant ovarian cancer using ultrasound responsive doxorubicin/curcumin co-deliver alginate nanodroplets. Colloids Surf. B Biointerfaces 2017, 153, 132–140.
  81. Sun, R.; Liu, X.; Li, G.; Wang, H.; Luo, Y.; Huang, G.; Wang, X.; Zeng, G.; Liu, Z.; Wu, S. Photoactivated H2 nanogenerator for enhanced chemotherapy of bladder cancer. ACS Nano 2020, 14, 8135–8148.
  82. Younis, M.; Faming, W.; Hongyan, Z.; Mengmeng, T.; Hang, S.; Liudi, Y. Iguratimod encapsulated PLGA-NPs improves therapeutic outcome in glioma, glioma stem-like cells and temozolomide resistant glioma cells. Nanomedicine 2019, 22, 102101.
  83. Chen, Y.; Zhang, M.; Jin, H.; Tang, Y.; Wu, A.; Xu, Q.; Huang, Y. Prodrug-like, PEGylated protein toxin trichosanthin for reversal of chemoresistance. Mol. Pharm. 2017, 14, 1429–1438.
  84. Chen, M.; Song, F.; Liu, Y.; Tian, J.; Liu, C.; Li, R.; Zhang, Q. A dual pH-sensitive liposomal system with charge-reversal and NO generation for overcoming multidrug resistance in cancer. Nanoscale 2019, 11, 3814–3826.
  85. Kommineni, N.; Mahira, S.; Domb, A.J.; Khan, W. Cabazitaxel-loaded nanocarriers for cancer therapy with reduced side effects. Pharmaceutics 2019, 11, 141.
  86. Li, X.; Wu, X.; Yang, H.; Li, L.; Ye, Z.; Rao, Y. A nuclear targeted Dox-aptamer loaded liposome delivery platform for the circumvention of drug resistance in breast cancer. Biomed. Pharmacother. 2019, 117, 109072.
  87. Zhang, L.; Su, H.; Liu, Y.; Pang, N.; Li, J.; Qi, X.R. Enhancing solid tumor therapy with sequential delivery of dexamethasone and docetaxel engineered in a single carrier to overcome stromal resistance to drug delivery. J. Control. Release 2019, 294, 1–16.
  88. Chen, Y.; Cheng, Y.; Zhao, P.; Zhang, S.; Li, M.; He, C.; Zhang, X.; Yang, T.; Yan, R.; Ye, P.; et al. Co-delivery of doxorubicin and imatinib by pH sensitive cleavable PEGylated nanoliposomes with folate-mediated targeting to overcome multidrug resistance. Int. J. Pharm. 2018, 542, 266–279.
  89. Zhu, Y.X.; Jia, H.R.; Duan, Q.Y.; Liu, X.; Yang, J.; Liu, Y.; Wu, F.G. Photosensitizer-doped and plasma membrane-responsive liposomes for nuclear drug delivery and multidrug resistance reversal. ACS Appl. Mater. Interfaces 2020, 12, 36882–36894.
  90. Wang, C.; Guan, W.; Peng, J.; Chen, Y.; Xu, G.; Dou, H. Gene/paclitaxel co-delivering nanocarriers prepared by framework-induced self-assembly for the inhibition of highly drug-resistant tumors. Acta Biomater. 2020, 103, 247–258.
  91. Wu, H.; Jin, H.; Wang, C.; Zhang, Z.; Ruan, H.; Sun, L.; Yang, C.; Li, Y.; Qin, W.; Wang, C. Synergistic cisplatin/doxorubicin combination chemotherapy for multidrug-resistant cancer via polymeric nanogels targeting delivery. ACS Appl. Mater. Interfaces 2017, 9, 9426–9436.
  92. Zhang, W.; Tung, C.H. Redox-responsive cisplatin nanogels for anticancer drug delivery. Chem. Commun. 2018, 54, 8367–8370.
  93. Zhang, X.; Chen, X.; Guo, Y.; Jia, H.R.; Jiang, Y.W.; Wu, F.G. Endosome/lysosome-detained supramolecular nanogels as an efflux retarder and autophagy inhibitor for repeated photodynamic therapy of multidrug-resistant cancer. Nanoscale Horiz. 2020, 5, 481–487.
  94. Sun, L.; Wang, D.; Chen, Y.; Wang, L.; Huang, P.; Li, Y.; Liu, Z.; Yao, H.; Shi, J. Core-shell hierarchical mesostructured silica nanoparticles for gene/chemo-synergetic stepwise therapy of multidrug-resistant cancer. Biomaterials 2017, 133, 219–228.
  95. Del Valle, A.C.; Yeh, C.K.; Huang, Y.F. Near infrared-activatable platinum-decorated gold nanostars for synergistic photothermal/ferroptotic therapy in combating cancer drug resistance. Adv. Healthc. Mater. 2020, 9, e2000864.
  96. Wang, Z.; Chang, Z.M.; Shao, D.; Zhang, F.; Chen, F.; Li, L.; Ge, M.F.; Hu, R.; Zheng, X.; Wang, Y.; et al. Janus gold triangle-mesoporous silica nanoplatforms for hypoxia-activated radio-chemo-photothermal therapy of liver cancer. ACS Appl. Mater. Interfaces 2019, 11, 34755–34765.
  97. Chen, J.; Jiang, Z.; Xu, W.; Sun, T.; Zhuang, X.; Ding, J.; Chen, X. Spatiotemporally targeted nanomedicine overcomes hypoxia-induced drug resistance of tumor cells after disrupting neovasculature. Nano Lett. 2020, 20, 6191–6198.
  98. Zhang, H.; Cui, W.; Qu, X.; Wu, H.; Qu, L.; Zhang, X.; Mäkilä, E.; Salonen, J.; Zhu, Y.; Yang, Z.; et al. Photothermal-responsive nanosized hybrid polymersome as versatile therapeutics codelivery nanovehicle for effective tumor suppression. Proc. Natl. Acad. Sci. USA 2019, 116, 7744–7749.
  99. Gao, J.; Wang, F.; Wang, S.; Liu, L.; Liu, K.; Ye, Y.; Wang, Z.; Wang, H.; Chen, B.; Jiang, J.; et al. Hyperthermia-triggered on-demand biomimetic nanocarriers for synergetic photothermal and chemotherapy. Adv. Sci. 2020, 7, 1903642.
  100. Yang, Q.; Xiao, Y.; Yin, Y.; Li, G.; Peng, J. Erythrocyte membrane-camouflaged IR780 and DTX coloading polymeric nanoparticles for imaging-guided cancer photo-chemo combination therapy. Mol. Pharm. 2019, 16, 3208–3220.
  101. Wang, J.; Lv, F.M.; Wang, D.L.; Du, J.L.; Guo, H.Y.; Chen, H.N.; Zhao, S.J.; Liu, Z.P.; Liu, Y. Synergistic antitumor effects on drug-resistant breast cancer of paclitaxel/lapatinib composite canocrystals. Molecules 2020, 25, 604.
  102. Li, L.; He, D.; Guo, Q.; Zhang, Z.; Ru, D.; Wang, L.; Gong, K.; Liu, F.; Duan, Y.; Li, H. Exosome-liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer. J. Nanobiotechnology 2022, 20, 50.
More