Chemoimmunotherapy combinations: translating basic knowledge into clinical successes

Chemoimmunotherapy combinations: translating basic knowledge into clinical successes

Play all audios:

Loading...

While chemotherapeutic agents were long solely associated with immunosuppression, clinical data demonstrate that the combination of some chemotherapies with immunomodulators can be


beneficial against cancer. Defining combinations featuring optimal anticancer activity along with minimal toxicity remains however a major challenge. Clinical evidence suggests that immune


responses in patients treated with combination therapies are associated with progression-free survival. Progress in understanding the mechanisms responsible for anticancer immune responses


following chemotherapy administration facilitated the translation of relevant chemoimmunotherapy combinations in the clinic. Almost 20 years ago Richard Lake and Bruce Robinson proposed that


combining chemotherapy and immunotherapy could result in therapeutic benefits [1]. This assumption notably relied on preclinical results obtained with the chemotherapy gemcitabine and


anti-CD40 antibody as an immunotherapy [2]. Chemotherapy or immunotherapy given alone failed to induce tumor control. However, treatment with gemcitabine followed by anti-CD40 drove tumor


regression in up to 80% of the mice and led to the development of immunological memory [2]. While these results demonstrated potential synergistic effects between chemotherapy and


immunomodulation, the clinical relevance of these observations for patients has long remained unexplored. Chemotherapy was indeed almost exclusively associated with immunosuppression because


of its ability to target cells dividing rapidly, including the cells of the immune system. Because of this, the effectiveness of chemotherapies was primarily attributed to their ability to


directly trigger cancer killing. When researchers in the early 70 s interrogated the mechanisms underlying the anticancer efficacy of two anthracyclines, adriamycin and daunorubicin, some


results seemed paradoxical. Daunorubicin featured enhanced activity against leukemic cells in vitro [3]. However, adriamycin featured better antileukemic activity than daunorubicin in vivo


despite the absence of major differences in biodistribution of the two drugs [4]. In addition, the superior anticancer effect of adriamycin over daunorubicin was lost in mice devoid of


immune responses due to previous irradiation [4], suggesting an involvement of immune responses in the anticancer activity of adriamycin. This contention was confirmed by multiple


investigators over the next fifty years. While high-dose chemotherapy induces immunosuppression [5], relevant doses of chemotherapy can trigger immune responses against cancer.


Chemotherapies drive cancer cell death. This process results in the emission of mediators that can be sensed by the immune system. We have for instance reported that the release of danger


signals such as High Mobility Group Box 1 (HMGB1) by dying tumor cells contributes to tumor immunogenicity [6]. HMGB1 binds to Toll-like receptor 4 (TLR4) on dendritic cells, ultimately


leading the activation of T cell-mediated anticancer immune responses [6]. The molecular mechanisms explaining the abilities of chemotherapies to trigger an immunogenic form of tumor cell


death (ICD) and their clinical relevance have been reviewed [7]. ICD is one of the ways by which chemotherapies can induce immune responses. Chemotherapies such as cyclophosphamide and


5-fluorouracil were notably shown to induce immune responses through their ability to eliminate regulatory T cells and myeloid derived suppressor cells [8], respectively. The mechanisms by


which chemotherapies unleash anticancer immune responses against cancer have been discussed [9, 10]. While tumor cells contribute to immunosuppression in the tumor microenvironment (TME),


activated immune cells can also paradoxically support the termination of anticancer immune responses. IFN-γ-producing CD8 T cells were shown to drive the expression of immunosuppressive


factors such as indoleamine-2,3-dioxygenase (IDO) and Programmed death-ligand 1 (PD-L1) in the TME [11]. The ability of T cell-derived IFN-γ to induce PD-L1 expression in cancer cells was


termed adaptive immune resistance [12]. Clinical observations in metastatic melanoma patients confirmed the relevance of this process by showing that patients responding to anti-PD-1


treatment featured enhanced proliferation of CD8 T cells in the TME [13]. Immune responses triggered by chemotherapies are rarely sufficient to drive tumor elimination, leading investigators


to interrogate the reasons accounting for tumor resistance to immunogenic chemotherapies. We and others have interrogated the relevance of tumor adaptive immune resistance following


chemotherapy administration. We showed that the release of IFN-γ drove the expression of PD-L1 on tumor cells, thereby leading to the dysfunction of PD-1-expressing T cells in tumors [14].


To restore productive anticancer immune responses, we combined anti-PD-1 treatment with chemotherapies. In two mouse models of colon cancer, we found that the addition of anti-PD-1 treatment


to a combined administration of the two chemotherapies 5-fluorouracil and oxaliplatin markedly enhanced T cell effector responses and mouse survival [14]. Similar results were obtained by


Pfirschke _et al_. who documented in lung, colon and fibrosarcoma cancer models that chemotherapies synergized with anti-PD-1 and anti-CTLA-4 therapy [15]. Overall, preclinical


investigations demonstrated the relevance of combined treatment of chemotherapy with immune checkpoint inhibition for durable cancer control (Fig. 1). The clinical relevance of combination


of chemotherapies and immunomodulation was recently explored in patients bearing MicroSatellite Stable (MSS) colon cancer. Building on early investigations described above suggesting


beneficial effects of combining 5-fluorouracil, oxaliplatin, and immunomodulation [14], Thibaudin et al. tested in a phase Ib/II clinical trial the safety and efficacy of a combination of


chemotherapy, with durvalumab, an anti-PD-L1 antibody and tremelimumab, an anti-CTLA-4 antibody administered to patients bearing metastatic colon cancer [16]. Results from 48 patients


revealed a median progression-free survival (PFS) of 8.2 months, which is superior to the expected median PFS of 5 to 6 months with chemotherapy alone. Importantly, 6 patients achieved a


complete response, indicating that chemo-immunotherapy combinations are clinically effective [16]. Analysis of immune responses against the tumor antigens telomerase and NY-ESO1 revealed


that the combination treatment induced enhanced T cell responses against these tumor antigens after one treatment cycle [16]. Importantly, increased T cell responses in this setting were


associated with longer PFS. While the combined treatment triggered adverse events in some of the patients leading to treatment discontinuation, these results altogether indicate that


combination therapies can harness anticancer immune responses in metastatic MSS colon cancer patients [16]. The example above is one of the successful implementations of chemotherapy and


immunomodulation combinations in the clinic (reviewed in [17]). These results are notably in line with recent findings documented in advanced oesophageal cancer. Immunomodulation using


anti-PD-1 antibodies administered with chemotherapy improves the overall survival of oesophageal squamous cell carcinoma (OSCC) patients as compared to chemotherapy alone [18]. A recent


phase 3 clinical trial interrogated the relevance of anti-PD-1 combined with different chemotherapy associations, oxaliplatin or cisplatin plus capecitabine or fluorouracil or paclitaxel,


compared to chemotherapies alone as first-line treatment for OSCC. Results revealed that the beneficial therapeutic effects of the combination were not restricted to the use of the cisplatin


and fluorouracil combination, but also to multiple chemotherapy combinations tested by the investigators [19]. These results not only provide additional therapeutic options for patients


suffering from oesophageal cancer, but also suggest that the ability to administer relevant clinically chemoimmunotherapy combinations is a concept that is broadly applicable (Fig. 1).


Further investigations into the genetic and immunological biomarkers associated with the success of chemoimmunotherapy combinations will ultimately ease further their clinical


implementation. REFERENCES * Lake RA, Robinson BW. Immunotherapy and chemotherapy-a practical partnership. Nat Rev Cancer. 2005;5:397–405. Article  CAS  PubMed  Google Scholar  * Nowak AK,


Robinson BW, Lake RA. Synergy between chemotherapy and immunotherapy in the treatment of established murine solid tumors. Cancer Res. 2003;63:4490–6. CAS  PubMed  Google Scholar  *


Meriwether WD, Bachur NR. Inhibition of DNA and RNA metabolism by daunorubicin and adriamycin in L1210 mouse leukemia. Cancer Res. 1972;32:1137–42. CAS  PubMed  Google Scholar  * Schwartz


HS, Grindey GB. Adriamycin and daunorubicin: a comparison of antitumor activities and tissue uptake in mice following immunosuppression. Cancer Res. 1973;33:1837–44. CAS  PubMed  Google


Scholar  * Hauser SL, Dawson DM, Lehrich JR, Beal MF, Kevy SV, Propper RD, et al. Intensive immunosuppression in progressive multiple sclerosis. A randomized, three-arm study of high-dose


intravenous cyclophosphamide, plasma exchange, and ACTH. N Engl J Med. 1983;308:173–80. Article  CAS  PubMed  Google Scholar  * Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C,


Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–9. Article  CAS  PubMed  Google Scholar


  * Sprooten J, Laureano RS, Vanmeerbeek I, Govaerts J, Naulaerts S, Borras DM, et al. Trial watch: chemotherapy-induced immunogenic cell death in oncology. Oncoimmunology. 2023;12:2219591.


Article  PubMed  PubMed Central  Google Scholar  * Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, et al. 5-Fluorouracil selectively kills tumor-associated


myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010;70:3052–61. Article  CAS  PubMed  Google Scholar  * Galluzzi L, Humeau J, Buque


A, Zitvogel L, Kroemer G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol. 2020;17:725–41. Article  PubMed  Google Scholar  * Dosset M,


Joseph EL, Rivera Vargas T, Apetoh L. Modulation of Determinant Factors to Improve Therapeutic Combinations with Immune Checkpoint Inhibitors. Cells. 2020;9:1727. Article  CAS  PubMed 


PubMed Central  Google Scholar  * Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, Gajewski TF. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is


driven by CD8(+) T cells. Sci Transl Med. 2013;5:200ra116. Article  PubMed  PubMed Central  Google Scholar  * Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev


Cancer. 2012;12:252–64. Article  CAS  PubMed  PubMed Central  Google Scholar  * Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by


inhibiting adaptive immune resistance. Nature. 2014;515:568–71. Article  CAS  PubMed  PubMed Central  Google Scholar  * Dosset M, Vargas TR, Lagrange A, Boidot R, Vegran F, Roussey A, et al.


PD-1/PD-L1 pathway: an adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology. 2018;7:e1433981. Article  PubMed  PubMed Central  Google


Scholar  * Pfirschke C, Engblom C, Rickelt S, Cortez-Retamozo V, Garris C, Pucci F, et al. Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity.


2016;44:343–54. Article  CAS  PubMed  PubMed Central  Google Scholar  * Thibaudin M, Fumet JD, Chibaudel B, Bennouna J, Borg C, Martin-Babau J, et al. First-line durvalumab and tremelimumab


with chemotherapy in RAS-mutated metastatic colorectal cancer: a phase 1b/2 trial. Nat Med. 2023;29:2087–98. Article  CAS  PubMed  PubMed Central  Google Scholar  * Rivera Vargas T, Apetoh


L. Can immunogenic chemotherapies relieve cancer cell resistance to immune checkpoint inhibitors? Front Immunol. 2019;10:1181. Article  PubMed  PubMed Central  Google Scholar  * Sun JM, Shen


L, Shah MA, Enzinger P, Adenis A, Doi T, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a


randomised, placebo-controlled, phase 3 study. Lancet. 2021;398:759–71. Article  CAS  PubMed  Google Scholar  * Xu J, Kato K, Raymond E, Hubner RA, Shu Y, Pan Y, et al. Tislelizumab plus


chemotherapy versus placebo plus chemotherapy as first-line treatment for advanced or metastatic oesophageal squamous cell carcinoma (RATIONALE-306): a global, randomised,


placebo-controlled, phase 3 study. Lancet Oncol. 2023;24:483–95. Article  CAS  PubMed  Google Scholar  Download references ACKNOWLEDGEMENTS This work was supported in part by the Brown


Center for Immunotherapy at Indiana University Melvin and Bren Simon Comprehensive Cancer Center. The figure was created with Biorender.com. AUTHOR INFORMATION AUTHORS AND AFFILIATIONS *


Brown Center for Immunotherapy, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA Lionel Apetoh


Authors * Lionel Apetoh View author publications You can also search for this author inPubMed Google Scholar CORRESPONDING AUTHOR Correspondence to Lionel Apetoh. ETHICS DECLARATIONS


COMPETING INTERESTS LA is a consultant for Brenus-Pharma. LA performed consultancy work for Roche, Merck, Bristol-Myers Squibb, and Orega Biotech and was a recipient of a research grant from


Sanofi. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. RIGHTS AND PERMISSIONS


Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Apetoh, L. Chemoimmunotherapy combinations: translating basic knowledge into clinical successes. _Genes Immun_ 25, 99–101


(2024). https://doi.org/10.1038/s41435-024-00264-9 Download citation * Published: 13 March 2024 * Issue Date: April 2024 * DOI: https://doi.org/10.1038/s41435-024-00264-9 SHARE THIS ARTICLE


Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided


by the Springer Nature SharedIt content-sharing initiative