Navigating the Immune Maze: Pioneering Strategies for Unshackling Cancer Immunotherapy Resistance
Abstract
:Simple Summary
Abstract
1. Introduction
2. The Immune Maze: Understanding the Complex Landscape
3. Frontline Foes: Decoding the Architects of Immunotherapy Resistance
4. Pioneering Strategies to Overcome Resistance
4.1. Combination Therapies
4.2. Tumor Microenvironment (TME)
4.3. Emerging Immune Checkpoints
4.4. Enhancing Immunotherapy with Oncolytic Viruses
4.5. Cell Therapy (ACT)
4.6. Cancer Vaccines
4.7. Navigating Medication-Induced Resistance in Immunotherapy
4.8. Integrated Strategies for Overcoming Resistance
5. Recent Insights and Developments in Overcoming Immunotherapy Resistance
5.1. Genetic Alterations and Immunotherapy Resistance
5.2. Epigenetic Dynamics and Their Role in Resistance
5.3. The Microbiome’s Influence on Immunotherapy Efficacy
6. Clinical Implications and Translational Approaches
7. Future Perspectives in Immunotherapy
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
ACT | adoptive cell therapy |
AI | artificial intelligence |
CAR | chimeric antigen receptor |
CTLs | cytotoxic T cells |
CTLA-4 | cytotoxic T-lymphocyte-associated antigen 4 |
cfDNA | cell-free DNA |
CTCs | circulating tumor cells |
EGFR | epidermal growth factor receptor |
LAG-3 | lymphocyte activation gene-3 |
MDSCs | myeloid-derived suppressor cells |
MHC | major histocompatibility complex |
NK | natural killer |
NSCLC | non-small-cell lung cancer |
PBMC | peripheral blood mononuclear cells |
PD-1 | programmed cell death protein 1 |
PD-L1 | programmed death-ligand 1 |
PDO | patient-derived organoids |
PDX | patient-derived xenograft |
TAMs | tumor-associated macrophages |
TAM1 | type-1 TAM |
TAM2 | type-2 TAM |
TAN1 | type-1 TAN |
TAN2 | type-2 TAN |
TANs | tumor-associated neutrophils |
TIGIT | T cell immunoreceptor with immunoglobulin and ITIM domain |
TIM-3 | T cell immunoglobulin and mucin-domain-containing-3 |
TGF | transforming growth factor |
Tregs | regulatory T cells |
TMB | tumor mutational burden |
TME | tumor microenvironment |
References
- Shin, Y.H.; Bang, S.; Park, S.M.; Ma, X.; Cassilly, C.; Graham, D.; Xavier, R.; Clardy, J. Revisiting Coley’s Toxins: Immunogenic Cardiolipins from Streptococcus pyogenes. J. Am. Chem. Soc. 2023, 145, 21183–21188. [Google Scholar] [CrossRef]
- McCarthy, E.F. The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthop. J. 2006, 26, 154–158. [Google Scholar]
- Coley, W.B. The treatment of malignant tumors by repeated inoculations of erysipelas; with a report of ten original cases. Am. J. Med. Sci. 1893, 105, 487–511. [Google Scholar] [CrossRef]
- Brunet, J.F.; Denizot, F.; Luciani, M.F.; Roux-Dosseto, M.; Suzan, M.; Mattei, M.G.; Golstein, P. A new member of the immunoglobulin superfamily—CTLA-4. Nature 1987, 328, 267–270. [Google Scholar] [CrossRef]
- Zahavi, D.; Weiner, L. Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.T.; Lee, S.H.; Heo, Y.S. Molecular Interactions of Antibody Drugs Targeting PD-1, PD-L1, and CTLA-4 in Immuno-Oncology. Molecules 2019, 24, 1190. [Google Scholar] [CrossRef]
- Yao, L.; Jia, G.; Lu, L.; Bao, Y.; Ma, W. Factors affecting tumor responders and predictive biomarkers of toxicities in cancer patients treated with immune checkpoint inhibitors. Int. Immunopharmacol. 2020, 85, 106628. [Google Scholar] [CrossRef]
- Bae, J.; Parayath, N.; Ma, W.; Amiji, M.; Munshi, N.; Anderson, K.C. BCMA peptide-engineered nanoparticles enhance induction and function of antigen-specific CD8(+) cytotoxic T lymphocytes against multiple myeloma: Clinical applications. Leukemia 2020, 34, 210–223. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Lu, L.; Ma, W. Efficacy, Safety, and Challenges of CAR T-Cells in the Treatment of Solid Tumors. Cancers 2022, 14, 5983. [Google Scholar] [CrossRef] [PubMed]
- Jogalekar, M.P.; Rajendran, R.L.; Khan, F.; Dmello, C.; Gangadaran, P.; Ahn, B.C. CAR T-Cell-Based gene therapy for cancers: New perspectives, challenges, and clinical developments. Front. Immunol. 2022, 13, 925985. [Google Scholar] [CrossRef] [PubMed]
- Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef]
- Li, X.; Zhang, S.; Guo, G.; Han, J.; Yu, J. Gut microbiome in modulating immune checkpoint inhibitors. EBioMedicine 2022, 82, 104163. [Google Scholar] [CrossRef] [PubMed]
- Jackson, C.M.; Choi, J.; Lim, M. Mechanisms of immunotherapy resistance: Lessons from glioblastoma. Nat. Immunol. 2019, 20, 1100–1109. [Google Scholar] [CrossRef] [PubMed]
- Khalaf, K.; Hana, D.; Chou, J.T.; Singh, C.; Mackiewicz, A.; Kaczmarek, M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front. Immunol. 2021, 12, 656364. [Google Scholar] [CrossRef] [PubMed]
- Haist, M.; Stege, H.; Grabbe, S.; Bros, M. The Functional Crosstalk between Myeloid-Derived Suppressor Cells and Regulatory T Cells within the Immunosuppressive Tumor Microenvironment. Cancers 2021, 13, 210. [Google Scholar] [CrossRef] [PubMed]
- Iliadi, C.; Verset, L.; Bouchart, C.; Martinive, P.; Van Gestel, D.; Krayem, M. The current understanding of the immune landscape relative to radiotherapy across tumor types. Front. Immunol. 2023, 14, 1148692. [Google Scholar] [CrossRef]
- Piper, M.; Kluger, H.; Ruppin, E.; Hu-Lieskovan, S. Immune Resistance Mechanisms and the Road to Personalized Immunotherapy. Am. Soc. Clin. Oncol. Educ. Book 2023, 43, e390290. [Google Scholar] [CrossRef]
- Fountzilas, E.; Tsimberidou, A.M.; Vo, H.H.; Kurzrock, R. Clinical trial design in the era of precision medicine. Genome Med. 2022, 14, 101. [Google Scholar] [CrossRef]
- Zhang, C.; Liu, X.; Jin, S.; Chen, Y.; Guo, R. Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef]
- Liao, J.; Li, X.; Gan, Y.; Han, S.; Rong, P.; Wang, W.; Li, W.; Zhou, L. Artificial intelligence assists precision medicine in cancer treatment. Front. Oncol. 2022, 12, 998222. [Google Scholar] [CrossRef]
- Brown, C.E.; Bucktrout, S.; Butterfield, L.H.; Futer, O.; Galanis, E.; Hormigo, A.; Lim, M.; Okada, H.; Prins, R.; Marr, S.S.; et al. The future of cancer immunotherapy for brain tumors: A collaborative workshop. J. Transl. Med. 2022, 20, 236. [Google Scholar] [CrossRef]
- Luo, J.; Li, X.; Wei, K.L.; Chen, G.; Xiong, D.D. Advances in the application of computational pathology in diagnosis, immunomicroenvironment recognition, and immunotherapy evaluation of breast cancer: A narrative review. J. Cancer Res. Clin. Oncol. 2023, 149, 12535–12542. [Google Scholar] [CrossRef] [PubMed]
- Abaza, A.; Sid Idris, F.; Anis Shaikh, H.; Vahora, I.; Moparthi, K.P.; Al Rushaidi, M.T.; Muddam, M.R.; Obajeun, O.A.; Jaramillo, A.P.; Khan, S. Programmed Cell Death Protein 1 (PD-1) and Programmed Cell Death Ligand 1 (PD-L1) Immunotherapy: A Promising Breakthrough in Cancer Therapeutics. Cureus 2023, 15, e44582. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Guo, Z.; Wang, L. Progress and Challenges of Immunotherapy Predictive Biomarkers for Triple Negative Breast Cancer in the Era of Single-Cell Multi-Omics. Life 2023, 13, 1189. [Google Scholar] [CrossRef] [PubMed]
- Lei, Z.N.; Tian, Q.; Teng, Q.X.; Wurpel, J.N.D.; Zeng, L.; Pan, Y.; Chen, Z.S. Understanding and targeting resistance mechanisms in cancer. MedComm (2020) 2023, 4, e265. [Google Scholar] [CrossRef]
- Kim, S.K.; Cho, S.W. The Evasion Mechanisms of Cancer Immunity and Drug Intervention in the Tumor Microenvironment. Front. Pharmacol. 2022, 13, 868695. [Google Scholar] [CrossRef] [PubMed]
- Dhatchinamoorthy, K.; Colbert, J.D.; Rock, K.L. Cancer Immune Evasion Through Loss of MHC Class I Antigen Presentation. Front. Immunol. 2021, 12, 636568. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Zhou, Z.; Dang, Q.; Xu, H.; Lv, J.; Li, H.; Han, X. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Theranostics 2022, 12, 6273–6290. [Google Scholar] [CrossRef]
- Tie, Y.; Tang, F.; Wei, Y.Q.; Wei, X.W. Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J. Hematol. Oncol. 2022, 15, 61. [Google Scholar] [CrossRef]
- Trujillo, J.A.; Luke, J.J.; Zha, Y.; Segal, J.P.; Ritterhouse, L.L.; Spranger, S.; Matijevich, K.; Gajewski, T.F. Secondary resistance to immunotherapy associated with beta-catenin pathway activation or PTEN loss in metastatic melanoma. J. Immunother. Cancer 2019, 7, 295. [Google Scholar] [CrossRef]
- Cendrowicz, E.; Sas, Z.; Bremer, E.; Rygiel, T.P. The Role of Macrophages in Cancer Development and Therapy. Cancers 2021, 13, 1946. [Google Scholar] [CrossRef] [PubMed]
- Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 2020, 18, 59. [Google Scholar] [CrossRef] [PubMed]
- Chakravarthi, B.V.; Nepal, S.; Varambally, S. Genomic and Epigenomic Alterations in Cancer. Am. J. Pathol. 2016, 186, 1724–1735. [Google Scholar] [CrossRef] [PubMed]
- Dai, E.; Zhu, Z.; Wahed, S.; Qu, Z.; Storkus, W.J.; Guo, Z.S. Epigenetic modulation of antitumor immunity for improved cancer immunotherapy. Mol. Cancer 2021, 20, 171. [Google Scholar] [CrossRef]
- Martinez-Jimenez, F.; Priestley, P.; Shale, C.; Baber, J.; Rozemuller, E.; Cuppen, E. Genetic immune escape landscape in primary and metastatic cancer. Nat. Genet. 2023, 55, 820–831. [Google Scholar] [CrossRef]
- Cao, J.; Yan, Q. Cancer Epigenetics, Tumor Immunity, and Immunotherapy. Trends Cancer 2020, 6, 580–592. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.; Turcan, S. Epigenetic Drugs and Their Immune Modulating Potential in Cancers. Biomedicines 2022, 10, 211. [Google Scholar] [CrossRef]
- Yang, J.; Xu, J.; Wang, W.; Zhang, B.; Yu, X.; Shi, S. Epigenetic regulation in the tumor microenvironment: Molecular mechanisms and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 210. [Google Scholar] [CrossRef]
- Lindau, D.; Gielen, P.; Kroesen, M.; Wesseling, P.; Adema, G.J. The immunosuppressive tumour network: Myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 2013, 138, 105–115. [Google Scholar] [CrossRef]
- Ma, T.; Renz, B.W.; Ilmer, M.; Koch, D.; Yang, Y.; Werner, J.; Bazhin, A.V. Myeloid-Derived Suppressor Cells in Solid Tumors. Cells 2022, 11, 310. [Google Scholar] [CrossRef]
- Sun, R.; Zhao, H.; Gao, D.S.; Ni, A.; Li, H.; Chen, L.; Lu, X.; Chen, K.; Lu, B. Amphiregulin couples IL1RL1(+) regulatory T cells and cancer-associated fibroblasts to impede antitumor immunity. Sci. Adv. 2023, 9, eadd7399. [Google Scholar] [CrossRef]
- Shi, H.; Li, K.; Ni, Y.; Liang, X.; Zhao, X. Myeloid-Derived Suppressor Cells: Implications in the Resistance of Malignant Tumors to T Cell-Based Immunotherapy. Front. Cell Dev. Biol. 2021, 9, 707198. [Google Scholar] [CrossRef]
- Li, K.; Shi, H.; Zhang, B.; Ou, X.; Ma, Q.; Chen, Y.; Shu, P.; Li, D.; Wang, Y. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct. Target. Ther. 2021, 6, 362. [Google Scholar] [CrossRef]
- Kopecka, J.; Salaroglio, I.C.; Perez-Ruiz, E.; Sarmento-Ribeiro, A.B.; Saponara, S.; De Las Rivas, J.; Riganti, C. Hypoxia as a driver of resistance to immunotherapy. Drug Resist. Updat. 2021, 59, 100787. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Zhao, Q.; Zhang, Y.; Liu, Z.; Zheng, Z.; Liu, S.; Meng, L.; Xin, Y.; Jiang, X. Targeting hypoxia in the tumor microenvironment: A potential strategy to improve cancer immunotherapy. J. Exp. Clin. Cancer Res. 2021, 40, 24. [Google Scholar] [CrossRef] [PubMed]
- Shi, T.; Zhu, J.; Zhang, X.; Mao, X. The Role of Hypoxia and Cancer Stem Cells in Development of Glioblastoma. Cancers 2023, 15, 2613. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharya, S.; Calar, K.; de la Puente, P. Mimicking tumor hypoxia and tumor-immune interactions employing three-dimensional in vitro models. J. Exp. Clin. Cancer Res. 2020, 39, 75. [Google Scholar] [CrossRef] [PubMed]
- Shurin, M.R.; Umansky, V. Cross-talk between HIF and PD-1/PD-L1 pathways in carcinogenesis and therapy. J. Clin. Investig. 2022, 132, e159473. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.; You, L.; Nepovimova, E.; Heger, Z.; Wu, W.; Kuca, K.; Adam, V. Hypoxia-inducible factors: Master regulators of hypoxic tumor immune escape. J. Hematol. Oncol. 2022, 15, 77. [Google Scholar] [CrossRef]
- Li, H.; Shao, S.; Cai, J.; Burner, D.; Lu, L.; Chen, Q.; Minev, B.; Ma, W. Artificial human antigen-presenting cells are superior to dendritic cells at inducing cytotoxic T-cell responses. Immunology 2017, 152, 462–471. [Google Scholar] [CrossRef] [PubMed]
- Lu, L.; Ma, W.; Johnson, C.H.; Khan, S.A.; Irwin, M.L.; Pusztai, L. In silico designed mRNA vaccines targeting CA-125 neoantigen in breast and ovarian cancer. Vaccine 2023, 41, 2073–2083. [Google Scholar] [CrossRef]
- Ma, W.; Smith, T.; Bogin, V.; Zhang, Y.; Ozkan, C.; Ozkan, M.; Hayden, M.; Schroter, S.; Carrier, E.; Messmer, D.; et al. Enhanced presentation of MHC class Ia, Ib and class II-restricted peptides encapsulated in biodegradable nanoparticles: A promising strategy for tumor immunotherapy. J. Transl. Med. 2011, 9, 34. [Google Scholar] [CrossRef] [PubMed]
- Kallingal, A.; Olszewski, M.; Maciejewska, N.; Brankiewicz, W.; Baginski, M. Cancer immune escape: The role of antigen presentation machinery. J. Cancer Res. Clin. Oncol. 2023, 149, 8131–8141. [Google Scholar] [CrossRef] [PubMed]
- Haddad, A.F.; Young, J.S.; Gill, S.; Aghi, M.K. Resistance to immune checkpoint blockade: Mechanisms, counter-acting approaches, and future directions. Semin. Cancer Biol. 2022, 86, 532–541. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Han, Y.; Zhang, Y.; Zhao, Q.; Wang, H.; Wei, J.; Meng, L.; Xin, Y.; Jiang, X. Overcoming acquired resistance to cancer immune checkpoint therapy: Potential strategies based on molecular mechanisms. Cell Biosci. 2023, 13, 120. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef] [PubMed]
- Marin-Acevedo, J.A.; Dholaria, B.; Soyano, A.E.; Knutson, K.L.; Chumsri, S.; Lou, Y. Next generation of immune checkpoint therapy in cancer: New developments and challenges. J. Hematol. Oncol. 2018, 11, 39. [Google Scholar] [CrossRef]
- Naimi, A.; Mohammed, R.N.; Raji, A.; Chupradit, S.; Yumashev, A.V.; Suksatan, W.; Shalaby, M.N.; Thangavelu, L.; Kamrava, S.; Shomali, N.; et al. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun. Signal. 2022, 20, 44. [Google Scholar] [CrossRef]
- Webb, E.S.; Liu, P.; Baleeiro, R.; Lemoine, N.R.; Yuan, M.; Wang, Y.H. Immune checkpoint inhibitors in cancer therapy. J. Biomed. Res. 2018, 32, 317–326. [Google Scholar] [CrossRef]
- Xie, Q.; Zhang, P.; Wang, Y.; Mei, W.; Zeng, C. Overcoming resistance to immune checkpoint inhibitors in hepatocellular carcinoma: Challenges and opportunities. Front. Oncol. 2022, 12, 958720. [Google Scholar] [CrossRef]
- Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 147–164. [Google Scholar] [CrossRef]
- Passaro, A.; Brahmer, J.; Antonia, S.; Mok, T.; Peters, S. Managing Resistance to Immune Checkpoint Inhibitors in Lung Cancer: Treatment and Novel Strategies. J. Clin. Oncol. 2022, 40, 598–610. [Google Scholar] [CrossRef] [PubMed]
- Metropulos, A.E.; Munshi, H.G.; Principe, D.R. The difficulty in translating the preclinical success of combined TGFbeta and immune checkpoint inhibition to clinical trial. EBioMedicine 2022, 86, 104380. [Google Scholar] [CrossRef]
- Tiwari, A.; Trivedi, R.; Lin, S.Y. Tumor microenvironment: Barrier or opportunity towards effective cancer therapy. J. Biomed. Sci. 2022, 29, 83. [Google Scholar] [CrossRef]
- Chen, M.L.; Pittet, M.J.; Gorelik, L.; Flavell, R.A.; Weissleder, R.; von Boehmer, H.; Khazaie, K. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-beta signals in vivo. Proc. Natl. Acad. Sci. USA 2005, 102, 419–424. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Guo, Y.; Liu, S.; Wang, H.; Zhu, J.; Ou, L.; Xu, X. Targeting regulatory T cells for immunotherapy in melanoma. Mol. Biomed. 2021, 2, 11. [Google Scholar] [CrossRef] [PubMed]
- Itahashi, K.; Irie, T.; Nishikawa, H. Regulatory T-cell development in the tumor microenvironment. Eur. J. Immunol. 2022, 52, 1216–1227. [Google Scholar] [CrossRef]
- Nishikawa, H.; Koyama, S. Mechanisms of regulatory T cell infiltration in tumors: Implications for innovative immune precision therapies. J. Immunother. Cancer 2021, 9, e002591. [Google Scholar] [CrossRef]
- Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef]
- Wang, S.; Zhao, X.; Wu, S.; Cui, D.; Xu, Z. Myeloid-derived suppressor cells: Key immunosuppressive regulators and therapeutic targets in hematological malignancies. Biomark. Res. 2023, 11, 34. [Google Scholar] [CrossRef]
- Zalfa, C.; Paust, S. Natural Killer Cell Interactions with Myeloid Derived Suppressor Cells in the Tumor Microenvironment and Implications for Cancer Immunotherapy. Front. Immunol. 2021, 12, 633205. [Google Scholar] [CrossRef]
- Jakos, T.; Pislar, A.; Jewett, A.; Kos, J. Myeloid-Derived Suppressor Cells Hamper Natural Killer Cell Activity in Cancer: Role of Peptidases. Crit. Rev. Immunol. 2021, 41, 77–99. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; He, H.; Jihu, R.; Zhou, J.; Zeng, R.; Yan, H. Novel Characterization of Myeloid-Derived Suppressor Cells in Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 698532. [Google Scholar] [CrossRef] [PubMed]
- Mehdizadeh, R.; Shariatpanahi, S.P.; Goliaei, B.; Ruegg, C. Targeting myeloid-derived suppressor cells in combination with tumor cell vaccination predicts anti-tumor immunity and breast cancer dormancy: An in silico experiment. Sci. Rep. 2023, 13, 5875. [Google Scholar] [CrossRef] [PubMed]
- He, Z.; Zhang, S. Tumor-Associated Macrophages and Their Functional Transformation in the Hypoxic Tumor Microenvironment. Front. Immunol. 2021, 12, 741305. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Saeed, A.; Liu, Q.; Jiang, Q.; Xu, H.; Xiao, G.G.; Rao, L.; Duo, Y. Macrophages in immunoregulation and therapeutics. Signal Transduct. Target. Ther. 2023, 8, 207. [Google Scholar] [CrossRef]
- Li, M.; He, L.; Zhu, J.; Zhang, P.; Liang, S. Targeting tumor-associated macrophages for cancer treatment. Cell Biosci. 2022, 12, 85. [Google Scholar] [CrossRef] [PubMed]
- Mehta, A.K.; Kadel, S.; Townsend, M.G.; Oliwa, M.; Guerriero, J.L. Macrophage Biology and Mechanisms of Immune Suppression in Breast Cancer. Front. Immunol. 2021, 12, 643771. [Google Scholar] [CrossRef]
- Feng, Y.; Ye, Z.; Song, F.; He, Y.; Liu, J. The Role of TAMs in Tumor Microenvironment and New Research Progress. Stem Cells Int. 2022, 2022, 5775696. [Google Scholar] [CrossRef]
- Yan, M.; Zheng, M.; Niu, R.; Yang, X.; Tian, S.; Fan, L.; Li, Y.; Zhang, S. Roles of tumor-associated neutrophils in tumor metastasis and its clinical applications. Front. Cell Dev. Biol. 2022, 10, 938289. [Google Scholar] [CrossRef]
- Que, H.; Fu, Q.; Lan, T.; Tian, X.; Wei, X. Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188762. [Google Scholar] [CrossRef] [PubMed]
- Wicks, E.E.; Semenza, G.L. Hypoxia-inducible factors: Cancer progression and clinical translation. J. Clin. Investig. 2022, 132, e159839. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Zhao, L.; Li, X.F. Hypoxia and the Tumor Microenvironment. Technol. Cancer Res. Treat. 2021, 20, 15330338211036304. [Google Scholar] [CrossRef]
- Patsoukis, N.; Wang, Q.; Strauss, L.; Boussiotis, V.A. Revisiting the PD-1 pathway. Sci. Adv. 2020, 6, eabd2712. [Google Scholar] [CrossRef]
- Yi, M.; Jiao, D.; Xu, H.; Liu, Q.; Zhao, W.; Han, X.; Wu, K. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol. Cancer 2018, 17, 129. [Google Scholar] [CrossRef]
- Zhang, Y.; Coleman, M.; Brekken, R.A. Perspectives on Hypoxia Signaling in Tumor Stroma. Cancers 2021, 13, 3070. [Google Scholar] [CrossRef] [PubMed]
- Shklovskaya, E.; Rizos, H. MHC Class I Deficiency in Solid Tumors and Therapeutic Strategies to Overcome It. Int. J. Mol. Sci. 2021, 22, 6741. [Google Scholar] [CrossRef]
- Wen, M.; Li, Y.; Qin, X.; Qin, B.; Wang, Q. Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota. Cells 2023, 12, 1882. [Google Scholar] [CrossRef]
- Zhou, J.; Bashey, A.; Zhong, R.; Corringham, S.; Messer, K.; Pu, M.; Ma, W.; Chut, T.; Soiffer, R.; Mitrovich, R.C.; et al. CTLA-4 blockade following relapse of malignancy after allogeneic stem cell transplantation is associated with T cell activation but not with increased levels of T regulatory cells. Biol. Blood Marrow Transplant. 2011, 17, 682–692. [Google Scholar] [CrossRef]
- Chen, X.; Zhang, W.; Yang, W.; Zhou, M.; Liu, F. Acquired resistance for immune checkpoint inhibitors in cancer immunotherapy: Challenges and prospects. Aging 2022, 14, 1048–1064. [Google Scholar] [CrossRef]
- Said, S.S.; Ibrahim, W.N. Cancer Resistance to Immunotherapy: Comprehensive Insights with Future Perspectives. Pharmaceutics 2023, 15, 1143. [Google Scholar] [CrossRef]
- Lao, Y.; Shen, D.; Zhang, W.; He, R.; Jiang, M. Immune Checkpoint Inhibitors in Cancer Therapy—How to Overcome Drug Resistance? Cancers 2022, 14, 3575. [Google Scholar] [CrossRef]
- Baxter, M.A.; Middleton, F.; Cagney, H.P.; Petty, R.D. Resistance to immune checkpoint inhibitors in advanced gastro-oesophageal cancers. Br. J. Cancer 2021, 125, 1068–1079. [Google Scholar] [CrossRef]
- Dutta, S.; Ganguly, A.; Chatterjee, K.; Spada, S.; Mukherjee, S. Targets of Immune Escape Mechanisms in Cancer: Basis for Development and Evolution of Cancer Immune Checkpoint Inhibitors. Biology 2023, 12, 218. [Google Scholar] [CrossRef] [PubMed]
- Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251. [Google Scholar] [CrossRef] [PubMed]
- Emran, T.B.; Shahriar, A.; Mahmud, A.R.; Rahman, T.; Abir, M.H.; Siddiquee, M.F.; Ahmed, H.; Rahman, N.; Nainu, F.; Wahyudin, E.; et al. Multidrug Resistance in Cancer: Understanding Molecular Mechanisms, Immunoprevention and Therapeutic Approaches. Front. Oncol. 2022, 12, 891652. [Google Scholar] [CrossRef] [PubMed]
- Flies, D.B.; Langermann, S.; Jensen, C.; Karsdal, M.A.; Willumsen, N. Regulation of tumor immunity and immunotherapy by the tumor collagen extracellular matrix. Front. Immunol. 2023, 14, 1199513. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Liu, X.; Chang, C.Y.; Wang, H.Y.; Wang, R.F. The Interplay between T Cells and Cancer: The Basis of Immunotherapy. Genes 2023, 14, 1008. [Google Scholar] [CrossRef]
- Mantovani, A.; Allavena, P.; Marchesi, F.; Garlanda, C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 2022, 21, 799–820. [Google Scholar] [CrossRef]
- Park, K.; Veena, M.S.; Shin, D.S. Key Players of the Immunosuppressive Tumor Microenvironment and Emerging Therapeutic Strategies. Front. Cell Dev. Biol. 2022, 10, 830208. [Google Scholar] [CrossRef]
- Wang, Y.; Huang, T.; Gu, J.; Lu, L. Targeting the metabolism of tumor-infiltrating regulatory T cells. Trends Immunol. 2023, 44, 598–612. [Google Scholar] [CrossRef]
- Bejarano, L.; Jordao, M.J.C.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
- Lee, J.B.; Ha, S.J.; Kim, H.R. Clinical Insights into Novel Immune Checkpoint Inhibitors. Front. Pharmacol. 2021, 12, 681320. [Google Scholar] [CrossRef]
- Dulal, D.; Boring, A.; Terrero, D.; Johnson, T.; Tiwari, A.K.; Raman, D. Tackling of Immunorefractory Tumors by Targeting Alternative Immune Checkpoints. Cancers 2023, 15, 2774. [Google Scholar] [CrossRef]
- Zhu, X.; Fan, C.; Xiong, Z.; Chen, M.; Li, Z.; Tao, T.; Liu, X. Development and application of oncolytic viruses as the nemesis of tumor cells. Front. Microbiol. 2023, 14, 1188526. [Google Scholar] [CrossRef]
- Chesney, J.A.; Ribas, A.; Long, G.V.; Kirkwood, J.M.; Dummer, R.; Puzanov, I.; Hoeller, C.; Gajewski, T.F.; Gutzmer, R.; Rutkowski, P.; et al. Randomized, Double-Blind, Placebo-Controlled, Global Phase III Trial of Talimogene Laherparepvec Combined with Pembrolizumab for Advanced Melanoma. J. Clin. Oncol. 2023, 41, 528–540. [Google Scholar] [CrossRef] [PubMed]
- Feldman, S.A.; Assadipour, Y.; Kriley, I.; Goff, S.L.; Rosenberg, S.A. Adoptive Cell Therapy—Tumor-Infiltrating Lymphocytes, T-Cell Receptors, and Chimeric Antigen Receptors. Semin. Oncol. 2015, 42, 626–639. [Google Scholar] [CrossRef]
- Li, J.; Xiao, Z.; Wang, D.; Jia, L.; Nie, S.; Zeng, X.; Hu, W. The screening, identification, design and clinical application of tumor-specific neoantigens for TCR-T cells. Mol. Cancer 2023, 22, 141. [Google Scholar] [CrossRef] [PubMed]
- Ingram, Z.; Madan, S.; Merchant, J.; Carter, Z.; Gordon, Z.; Carey, G.; Webb, T.J. Targeting Natural Killer T Cells in Solid Malignancies. Cells 2021, 10, 1329. [Google Scholar] [CrossRef] [PubMed]
- Zhu, S.; Zhang, T.; Zheng, L.; Liu, H.; Song, W.; Liu, D.; Li, Z.; Pan, C.X. Combination strategies to maximize the benefits of cancer immunotherapy. J. Hematol. Oncol. 2021, 14, 156. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef] [PubMed]
- Kaczmarek, M.; Poznanska, J.; Fechner, F.; Michalska, N.; Paszkowska, S.; Napierala, A.; Mackiewicz, A. Cancer Vaccine Therapeutics: Limitations and Effectiveness—A Literature Review. Cells 2023, 12, 2159. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.W.; Yam, J.W.P.; Mao, X. Dendritic Cell Vaccines: A Shift from Conventional Approach to New Generations. Cells 2023, 12, 2147. [Google Scholar] [CrossRef] [PubMed]
- Gupta, M.; Wahi, A.; Sharma, P.; Nagpal, R.; Raina, N.; Kaurav, M.; Bhattacharya, J.; Rodrigues Oliveira, S.M.; Dolma, K.G.; Paul, A.K.; et al. Recent Advances in Cancer Vaccines: Challenges, Achievements, and Futuristic Prospects. Vaccines 2022, 10, 2011. [Google Scholar] [CrossRef] [PubMed]
- Goodman, R.S.; Johnson, D.B.; Balko, J.M. Corticosteroids and Cancer Immunotherapy. Clin. Cancer Res. 2023, 29, 2580–2587. [Google Scholar] [CrossRef] [PubMed]
- Kalfeist, L.; Galland, L.; Ledys, F.; Ghiringhelli, F.; Limagne, E.; Ladoire, S. Impact of Glucocorticoid Use in Oncology in the Immunotherapy Era. Cells 2022, 11, 770. [Google Scholar] [CrossRef]
- Meng, L.; Wei, Y.; Xiao, Y. Chemo-immunoablation of solid tumors: A new concept in tumor ablation. Front. Immunol. 2022, 13, 1057535. [Google Scholar] [CrossRef]
- Li, J.Y.; Chen, Y.P.; Li, Y.Q.; Liu, N.; Ma, J. Chemotherapeutic and targeted agents can modulate the tumor microenvironment and increase the efficacy of immune checkpoint blockades. Mol. Cancer 2021, 20, 27. [Google Scholar] [CrossRef]
- Eng, L.; Sutradhar, R.; Niu, Y.; Liu, N.; Liu, Y.; Kaliwal, Y.; Powis, M.L.; Liu, G.; Peppercorn, J.M.; Bedard, P.L.; et al. Impact of Antibiotic Exposure before Immune Checkpoint Inhibitor Treatment on Overall Survival in Older Adults with Cancer: A Population-Based Study. J. Clin. Oncol. 2023, 41, 3122–3134. [Google Scholar] [CrossRef]
- Peng, C.; Rabold, K.; Mulder, W.J.M.; Jaeger, M.; Netea-Maier, R.T. Kinase Inhibitors’ Effects on Innate Immunity in Solid Cancers. Cancers 2021, 13, 5695. [Google Scholar] [CrossRef]
- Castelo-Soccio, L.; Kim, H.; Gadina, M.; Schwartzberg, P.L.; Laurence, A.; O’Shea, J.J. Protein kinases: Drug targets for immunological disorders. Nat. Rev. Immunol. 2023, 23, 787–806. [Google Scholar] [CrossRef]
- Fogli, L.K.; Aurigemma, R.; Sommers, C.L.; Singh, A.; Bourcier, K.; Ernstoff, M.S.; NCI Cell Therapy Workshop Committee. Challenges and next steps in the advancement of immunotherapy: Summary of the 2018 and 2020 National Cancer Institute workshops on cell-based immunotherapy for solid tumors. J. Immuno Ther. Cancer 2021, 9, e003048. [Google Scholar] [CrossRef]
- Xu, S.; Tan, S.; Guo, L. Patient-Derived Organoids as a Promising Tool for Multimodal Management of Sarcomas. Cancers 2023, 15, 4339. [Google Scholar] [CrossRef] [PubMed]
- Seyhan, A.A.; Carini, C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int. J. Mol. Sci. 2022, 24, 41. [Google Scholar] [CrossRef] [PubMed]
- Bai, R.; Lv, Z.; Xu, D.; Cui, J. Predictive biomarkers for cancer immunotherapy with immune checkpoint inhibitors. Biomark. Res. 2020, 8, 34. [Google Scholar] [CrossRef]
- Shao, J.; Jin, Y.; Jin, C. A new approach to overcoming resistance to immunotherapy: Nanotechnology. Front. Oncol. 2023, 13, 1210245. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Zhang, T.; Xia, R.; Wei, Y.; Wei, X. Targeting the tumor stroma for cancer therapy. Mol. Cancer 2022, 21, 208. [Google Scholar] [CrossRef]
- Curigliano, G.; Gelderblom, H.; Mach, N.; Doi, T.; Tai, D.; Forde, P.M.; Sarantopoulos, J.; Bedard, P.L.; Lin, C.C.; Hodi, F.S.; et al. Phase I/Ib Clinical Trial of Sabatolimab, an Anti-TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti-PD-1 Antibody, in Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 3620–3629. [Google Scholar] [CrossRef]
- Finck, A.V.; Blanchard, T.; Roselle, C.P.; Golinelli, G.; June, C.H. Engineered cellular immunotherapies in cancer and beyond. Nat. Med. 2022, 28, 678–689. [Google Scholar] [CrossRef]
- Kiaie, S.H.; Salehi-Shadkami, H.; Sanaei, M.J.; Azizi, M.; Shokrollahi Barough, M.; Nasr, M.S.; Sheibani, M. Nano-immunotherapy: Overcoming delivery challenge of immune checkpoint therapy. J. Nanobiotechnol. 2023, 21, 339. [Google Scholar] [CrossRef]
- Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front. Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef] [PubMed]
- Audia, J.E.; Campbell, R.M. Histone Modifications and Cancer. Cold Spring Harb. Perspect. Biol. 2016, 8, a019521. [Google Scholar] [CrossRef]
- Xu, J.; Xu, H.M.; Yang, M.F.; Liang, Y.J.; Peng, Q.Z.; Zhang, Y.; Tian, C.M.; Wang, L.S.; Yao, J.; Nie, Y.Q.; et al. New Insights Into the Epigenetic Regulation of Inflammatory Bowel Disease. Front. Pharmacol. 2022, 13, 813659. [Google Scholar] [CrossRef]
- Wang, C.; Wang, Z.; Yao, T.; Zhou, J.; Wang, Z. The immune-related role of beta-2-microglobulin in melanoma. Front. Oncol. 2022, 12, 944722. [Google Scholar] [CrossRef] [PubMed]
- Liu, F.; Zhong, F.; Wu, H.; Che, K.; Shi, J.; Wu, N.; Fu, Y.; Wang, Y.; Hu, J.; Qian, X.; et al. Prevalence and Associations of Beta2-Microglobulin Mutations in MSI-H/dMMR Cancers. Oncologist 2023, 28, e136–e144. [Google Scholar] [CrossRef]
- Hu, Q.; Bian, Q.; Rong, D.; Wang, L.; Song, J.; Huang, H.S.; Zeng, J.; Mei, J.; Wang, P.Y. JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens. Front. Bioeng. Biotechnol. 2023, 11, 1110765. [Google Scholar] [CrossRef]
- Shen, H.; Huang, F.; Zhang, X.; Ojo, O.A.; Li, Y.; Trummell, H.Q.; Anderson, J.C.; Fiveash, J.; Bredel, M.; Yang, E.S.; et al. Selective suppression of melanoma lacking IFN-gamma pathway by JAK inhibition depends on T cells and host TNF signaling. Nat. Commun. 2022, 13, 5013. [Google Scholar] [CrossRef]
- Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising targets for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 9. [Google Scholar] [CrossRef]
- Blass, E.; Ott, P.A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 2021, 18, 215–229. [Google Scholar] [CrossRef]
- Mehdi, A.; Rabbani, S.A. Role of Methylation in Pro- and Anti-Cancer Immunity. Cancers 2021, 13, 545. [Google Scholar] [CrossRef]
- Desaulniers, D.; Vasseur, P.; Jacobs, A.; Aguila, M.C.; Ertych, N.; Jacobs, M.N. Integration of Epigenetic Mechanisms into Non-Genotoxic Carcinogenicity Hazard Assessment: Focus on DNA Methylation and Histone Modifications. Int. J. Mol. Sci. 2021, 22, 10969. [Google Scholar] [CrossRef]
- Markouli, M.; Strepkos, D.; Basdra, E.K.; Papavassiliou, A.G.; Piperi, C. Prominent Role of Histone Modifications in the Regulation of Tumor Metastasis. Int. J. Mol. Sci. 2021, 22, 2778. [Google Scholar] [CrossRef]
- Xiong, D.; Zhang, L.; Sun, Z.J. Targeting the epigenome to reinvigorate T cells for cancer immunotherapy. Mil. Med. Res. 2023, 10, 59. [Google Scholar] [CrossRef]
- Hu, C.; Liu, X.; Zeng, Y.; Liu, J.; Wu, F. DNA methyltransferase inhibitors combination therapy for the treatment of solid tumor: Mechanism and clinical application. Clin. Epigenet. 2021, 13, 166. [Google Scholar] [CrossRef]
- Shen, C.; Li, M.; Duan, Y.; Jiang, X.; Hou, X.; Xue, F.; Zhang, Y.; Luo, Y. HDAC inhibitors enhance the anti-tumor effect of immunotherapies in hepatocellular carcinoma. Front. Immunol. 2023, 14, 1170207. [Google Scholar] [CrossRef]
- Lu, G.; Jin, S.; Lin, S.; Gong, Y.; Zhang, L.; Yang, J.; Mou, W.; Du, J. Update on histone deacetylase inhibitors in peripheral T-cell lymphoma (PTCL). Clin. Epigenet. 2023, 15, 124. [Google Scholar] [CrossRef]
- Derosa, L.; Routy, B.; Thomas, A.M.; Iebba, V.; Zalcman, G.; Friard, S.; Mazieres, J.; Audigier-Valette, C.; Moro-Sibilot, D.; Goldwasser, F.; et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat. Med. 2022, 28, 315–324. [Google Scholar] [CrossRef]
- Patel, P.; Poudel, A.; Kafle, S.; Thapa Magar, M.; Cancarevic, I. Influence of Microbiome and Antibiotics on the Efficacy of Immune Checkpoint Inhibitors. Cureus 2021, 13, e16829. [Google Scholar] [CrossRef]
- Najmi, M.; Tran, T.; Witt, R.G.; Nelson, K.C. Modulation of the Gut Microbiome to Enhance Immunotherapy Response in Metastatic Melanoma Patients: A Clinical Review. Dermatol. Ther. 2022, 12, 2489–2497. [Google Scholar] [CrossRef]
- Villemin, C.; Six, A.; Neville, B.A.; Lawley, T.D.; Robinson, M.J.; Bakdash, G. The heightened importance of the microbiome in cancer immunotherapy. Trends Immunol. 2023, 44, 44–59. [Google Scholar] [CrossRef]
- Sarhadi, V.K.; Armengol, G. Molecular Biomarkers in Cancer. Biomolecules 2022, 12, 1021. [Google Scholar] [CrossRef]
- Seyhan, A.A.; Carini, C. Are innovation and new technologies in precision medicine paving a new era in patients centric care? J. Transl. Med. 2019, 17, 114. [Google Scholar] [CrossRef]
- Wang, W.; Li, Y.; Lin, K.; Wang, X.; Tu, Y.; Zhuo, Z. Progress in building clinically relevant patient-derived tumor xenograft models for cancer research. Anim. Model. Exp. Med. 2023, 6, 381–398. [Google Scholar] [CrossRef]
- Chen, K.; Li, Y.; Wang, B.; Yan, X.; Tao, Y.; Song, W.; Xi, Z.; He, K.; Xia, Q. Patient-derived models facilitate precision medicine in liver cancer by remodeling cell-matrix interaction. Front. Immunol. 2023, 14, 1101324. [Google Scholar] [CrossRef]
- Chitrangi, S.; Vaity, P.; Jamdar, A.; Bhatt, S. Patient-derived organoids for precision oncology: A platform to facilitate clinical decision making. BMC Cancer 2023, 23, 689. [Google Scholar] [CrossRef]
- Singh, S.; Kumar, R.; Payra, S.; Singh, S.K. Artificial Intelligence and Machine Learning in Pharmacological Research: Bridging the Gap Between Data and Drug Discovery. Cureus 2023, 15, e44359. [Google Scholar] [CrossRef]
- Dlamini, Z.; Francies, F.Z.; Hull, R.; Marima, R. Artificial intelligence (AI) and big data in cancer and precision oncology. Comput. Struct. Biotechnol. J. 2020, 18, 2300–2311. [Google Scholar] [CrossRef]
- Subbiah, V. The next generation of evidence-based medicine. Nat. Med. 2023, 29, 49–58. [Google Scholar] [CrossRef]
- Chen, Q.; Jia, G.; Zhao, X.; Bao, Y.; Zhang, Y.; Ozkan, C.; Minev, B.; Ma, W. Novel Survivin Peptides Screened with Computer Algorithm Induce Cytotoxic T Lymphocytes with Higher Cytotoxic Efficiency to Cancer Cells. Front. Mol. Biosci. 2020, 7, 570003. [Google Scholar] [CrossRef]
- Chen, Q.; Bao, Y.; Burner, D.; Kaushal, S.; Zhang, Y.; Mendoza, T.; Bouvet, M.; Ozkan, C.; Minev, B.; Ma, W. Tumor growth inhibition by mSTEAP peptide nanovaccine inducing augmented CD8(+) T cell immune responses. Drug Deliv. Transl. Res. 2019, 9, 1095–1105. [Google Scholar] [CrossRef]
- Ma, W.; Chen, M.; Kaushal, S.; McElroy, M.; Zhang, Y.; Ozkan, C.; Bouvet, M.; Kruse, C.; Grotjahn, D.; Ichim, T.; et al. PLGA nanoparticle-mediated delivery of tumor antigenic peptides elicits effective immune responses. Int. J. Nanomed. 2012, 7, 1475–1487. [Google Scholar] [CrossRef] [PubMed]
- Haimi, M. The tragic paradoxical effect of telemedicine on healthcare disparities—A time for redemption: A narrative review. BMC Med. Inform. Decis. Mak. 2023, 23, 95. [Google Scholar] [CrossRef] [PubMed]
Strategies | Description | Key Components and Benefits | Representative Drugs/Cells/Vaccines | References |
---|---|---|---|---|
Combination Therapies | Integration of several therapeutic modalities to optimize oncological outcomes. | Synergistic modalities enhance response. Versatility against varying tumor behaviors. Potential for prolonged patient benefits. | Anti-NKG2A: Monalizumab, Anti-PD-1: Nivolumab, Pembrolizumab Anti-PD-L1: Atezolizumab, Avelumab, Anti-CTLA-4: Ipilimumab, Durvalumab | [94,95,96,110] |
TME | Considers the composite of stromal and immune cells intertwined with signaling pathways. Affects tumor progression and anti-tumor immunity. | Stroma including ECM and fibroblasts; mesenchymal stromal cells; and immune cells such as TAMs, TANs, and Tregs, signaling pathways that influence tumor progression. | Anti-LOXL2: Simtuzumab, anti-hyaluronic acid: PEGPH20, anti-CTGF: Pamrevlumab, anti-Integrin: Cilengitide, ATN-161, MEDI-522, anti-TGF-β: Fresolimumab, etc. | [97,98,127] |
Immune Checkpoints (ICIs) | Novel checkpoints open up promising therapeutic possibilities. They modulate immune functions. | Potential checkpoints like TIGIT, TIM-3, and LAG-3 receptors, expanding therapeutic avenues. | Anti-LAG-3 mAbs: Relatlimab, Favezelimab, REGN3767, GSK2831781, LAG525, TSR-033, Relatlimab + Nivolumab, etc. Anti-TIM3: Sabatolimab, spartalizumab. | [127,128] |
Adoptive Cell Therapy (ACT) | Capitalizes on an individual’s immune cells. Offers a tailored therapeutic approach. | Precision with techniques like TIL extraction; potential of CAR-T cells provide a tailored therapeutic approach. Enhanced therapeutic results when combined with other modalities. | Tumor-infiltrating lymphocytes (TILs), T cell-receptor-engineered T (TCR-T) cells, natural killer T (NKT) cells | [107,108,109] |
Cancer Vaccines | Utilization of neoantigens to boost immune responses targeting tumors. | Innovation with DC vaccines and viral vector vaccines; enhances immune response. | Peptide vaccines: Gardasil®, gp96, OSE2101, DSP-7888, etc.; DNA vaccines: HER2, VGX-3100, WT1, P, MA, hTERT, etc. mRNA vaccines: BNT112, BNT113, MAGE-A3, KRAS, etc.; virus-based vaccines: PROSTVAC-V/F, TG4010, BT-001; cell-based vaccines: DC vaccines; GVAX, etc. | [111,112,113,114] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yao, L.; Wang, Q.; Ma, W. Navigating the Immune Maze: Pioneering Strategies for Unshackling Cancer Immunotherapy Resistance. Cancers 2023, 15, 5857. https://doi.org/10.3390/cancers15245857
Yao L, Wang Q, Ma W. Navigating the Immune Maze: Pioneering Strategies for Unshackling Cancer Immunotherapy Resistance. Cancers. 2023; 15(24):5857. https://doi.org/10.3390/cancers15245857
Chicago/Turabian StyleYao, Liqin, Qingqing Wang, and Wenxue Ma. 2023. "Navigating the Immune Maze: Pioneering Strategies for Unshackling Cancer Immunotherapy Resistance" Cancers 15, no. 24: 5857. https://doi.org/10.3390/cancers15245857
APA StyleYao, L., Wang, Q., & Ma, W. (2023). Navigating the Immune Maze: Pioneering Strategies for Unshackling Cancer Immunotherapy Resistance. Cancers, 15(24), 5857. https://doi.org/10.3390/cancers15245857