750
Views
1
CrossRef citations to date
0
Altmetric
Introduction

Cancer Immunology and Immunotherapy: From Defining Basic Immunology to Leading the Fight Against Cancer

ABSTRACT

The past decade has seen the advent and widespread use of several immunotherapeutic modalities that have markedly improved treatment outcomes for patients with various cancers. Nevertheless, the study of cancer immunology traces its roots back to the inception of modern immunology, and played a critical role in the of discovery of central immunological concepts and development of key technologies and methodologies and that have propelled advances in all areas of immunology.

The past decade has seen the advent and widespread use of several immunotherapeutic modalities that have markedly improved treatment outcomes for patients with various cancers. Nevertheless, the study of cancer immunology traces its roots back to the inception of modern immunology, and played a critical role in the of discovery of central immunological concepts and development of key technologies and methodologies and that have propelled advances in all areas of immunology. For instance, the discovery by Kohler and Milstein that myeloma cells can be fused with immunized B cells to generate immortalized hybridomas that can perpetually secrete monoclonal antibodies that bind defined antigens enabled the development of a myriad of foundational basic investigatory, diagnostic, and therapeutic applications (Milstein Citation1999). Additionally, the seminal tumor transplantation studies by George Snell and colleagues using inbred and congenic mouse strains defined the major histocompatibility locus (MHC) (Snell Citation1981). This led to the subsequent discovery that binding of the T cell antigen receptor (TCR) to cognate antigen is restricted by self-MHC (Zinkernagel and Doherty Citation1974) (HLA in humans (Ryder et al. Citation1981)), which has been central to understanding immunity to infectious diseases, autoimmunity, allergy, transplantation tolerance, and of course the development of cancer immunotherapies.

Given that the immune system principally evolved to neutralize infectious agents, and for decades standard of care cancer treatments included chemotherapy, radiation, surgery, and hormonal blockade, it was not clear until recently that the basic study of cancer immunology would ultimately advance clinical oncology. Further, basic discoveries in tumor biology spurred the development of promising new therapies designed to directly kill tumors via non-immunological mechanisms, such as inhibitors that target either oncogenic mutated tyrosine kinases (Druker et al. Citation2006; Flaherty et al. Citation2010) or factors that promote tumor angiogenesis (Abdollahi and Folkman Citation2010). It has thus been remarkable that immunotherapies have not only significantly advanced the clinical treatment of cancer, but also that “non-immunological” therapies such as radiation (Sharabi et al. Citation2015), chemotherapy (Apetoh et al. Citation2007), hormonal blockade (Adler Citation2007), and oncogenic kinase inhibition (Knight et al. Citation2013) work in part by promoting anti-tumor immunity.

Immune responses to infections occur through the coordinated actions of innate immune cells that acquire pathogen-derived antigens while being activated by common pathogen-associated molecular patterns (PAMPs), which then program adaptive B and T cells to secrete pathogen-specific antibodies or cytokines and other effector molecules that promote the cytotoxic killing of infected cells, respectively. Importantly, T cells also have the ability to recognize MHC-complexed peptide fragments processed from mutated self-antigens (i.e., neoepitopes) that arise in tumors (Brennick et al. Citation2017; Lurquin et al. Citation1989; Tran et al. Citation2017). Nevertheless, given that tumors arise from self and hence do not express PAMPs, tumor-specific T cells typically fail to develop sufficient effector functionality to control tumors, and rather tend to be suppressed by the same tolerance mechanisms that limit the ability of self-reactive T cells to cause autoimmunity (Adler and Vella Citation2013). Hence, T cell-based cancer immunotherapies generally work by overcoming some aspect of tumor-specific T cell tolerance.

Dendritic cells that present pathogen-derived peptide antigens complexed to MHC prime cognate T cells to undergo expansion and effector differentiation (Mellman et al. Citation1998). When dendritic cells acquire and present tumor antigens, however, the absence of PAMPs as well as exposure to suppressive factors can cause them to adopt a tolerogenic phenotype where they program tumor-specific T cells to become dysfunctional (Mihalyo et al. Citation2007; Scarlett et al. Citation2012). Based on the ability to load dendritic cells with tumor antigens under ex vivo culture conditions that promote optimal T cell priming capacity (Palucka et al. Citation2007), in 2010 the dendritic cell-based vaccine Provenge (sipuleucel-T) designed to treat castration-resistant prostate cancer was the first therapeutic tumor vaccine to receive FDA approval (Kantoff et al. Citation2010). The thematic article by Fu and colleagues (Fu et al. Citation2022) provides an update on the continuing efforts to improve the efficacy and extend the range of dendritic cell-based vaccines to treat a variety of cancers.

Tumor growth sometimes causes sufficient inflammation to induce dendritic cell activation and hence priming of tumor-specific effector T cells that subsequently migrate into the tumors. Nevertheless, the repetitive contact with tumor antigen causes these T cells to lose their cytotoxic capacity and become functionally exhausted (Wherry and Kurachi Citation2015). Since these exhausted T cells express high levels of checkpoint receptors such as CTLA-4, PD-1, and LAG-3 that suppress effector functionality, monoclonal antibody (mAb) antagonists to these checkpoint receptors or their ligands (i.e., checkpoint blockers) can restore tumor-specific T cell function and elicit meaningful therapeutic responses in a subset of patients with melanoma and other solid tumors (Baumeister et al. Citation2016; Sharma and Allison Citation2015; Tawbi et al. Citation2022). The thematic article by Koguchi and Redmond (Koguchi and Redmond Citation2022) describe a potential on-treatment biomarker that could guide adjustments in treatment regimens that maximize therapeutic responses for individual patients receiving checkpoint blockers or other immunotherapeutic mAbs.

The efficacy of checkpoint therapy is in part limited because only a subset of exhausted T cells can be functionally rejuvenated (Ghoneim et al. Citation2017; Miller et al. Citation2019; Yost et al. Citation2019). Additionally, tumor-specific T cells typically only comprise a minor fraction of tumor-infiltrating lymphocytes (TIL) (Scheper et al. Citation2019; Simoni et al. Citation2018). Strategies are thus being explored to activate non-specific TIL, since they are more abundant but less exhausted (Caushi et al. Citation2021). The thematic article by Long and colleagues (Long et al. Citation2022) discusses recent advances in the development of bispecific antibodies that simultaneously bind a surface antigen on tumor cells as well as the TCR on T cells to enable tumor-non-specific TIL to directly kill tumor cells in an MHC-non-restricted manner.

Another therapeutic modality that enables MHC-non-restricted tumor cell killing utilizes engineered T cells with chimeric antigen receptors (CARs) that recognize a tumor cell surface antigen in its natural form. CAR T-cells have proven highly effective in treating B cell malignancies (Lim and June Citation2017), although it has been difficult to adapt them for treating solid tumors, in part due to the challenge of finding suitable tumor cell surface antigens that are not expressed at appreciable levels in essential healthy tissues. Conejo-Garcia and Guevara-Patino (Conejo-Garcia and Guevara-Patino Citation2022) discuss exciting advances on this front that may soon make CAR T-cell therapy effective in treating various solid tumors.

Preceding much of the discoveries regarding the mechanisms of anti-tumor immunity that fueled the development of the immunotherapies mentioned above, in the 1970s Bacillus Calmette-Guerin (BCG) became a standard first-line treatment for non-invasive bladder cancer. The final thematic article by Ward Grados and colleagues (Ward Grados et al. Citation2022) covers new developments in how BCG and other immunotherapies are being combined to further enhance therapeutic efficacy in patients with bladder cancer.

It was not possible in these five thematic reviews to fully cover the many exciting and innovative approaches that are currently being pursued to develop new and improve existing cancer immunotherapies. Nevertheless, we hope they convey that our rapidly expanding understanding of how immune function is regulated is providing a plethora of opportunities to develop varied therapeutic strategies to combat a disease that has itself evolved numerous means to escape natural mechanisms of tumor control.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Abdollahi A, Folkman J. 2010. Evading tumor evasion: current concepts and perspectives of anti-angiogenic cancer therapy. Drug Resist Updat. 13(1–2):16–28.
  • Adler AJ. 2007. Mechanisms of T cell tolerance and suppression in cancer mediated by tumor-associated antigens and hormones. Curr Cancer Drug Targets. 7(1):3–14.
  • Adler AJ, Vella AT. 2013. Betting on improved cancer immunotherapy by doubling down on CD134 and CD137 co-stimulation. Oncoimmunology. 2(1):e22837.
  • Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, et al. 2007. Toll-like receptor 4–dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 13(9):1050–59.
  • Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. 2016. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol. 34539-73(1):539–73.
  • Brennick CA, George MM, Corwin WL, Srivastava PK, Ebrahimi-Nik H. 2017. Neoepitopes as cancer immunotherapy targets: key challenges and opportunities. Immunotherapy. 9(4):361–71.
  • Caushi JX, Zhang J, Ji Z, Vaghasia A, Zhang B, Hsiue EH, Mog BJ, Hou W, Justesen S, Blosser R, et al. 2021. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature. 596(7870):126–32. doi:10.1038/s41586-021-03752-4.
  • Conejo-Garcia JR, Guevara-Patino JA. 2022. Barriers and opportunities for CAR T-cell targeting of solid tumors. Immunol Invest. 1–11. doi:10.1080/08820139.2022.2096463
  • Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM, et al. 2006. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 355(23):2408–17.
  • Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O’Dwyer PJ, Lee RJ, Grippo JF, Nolop K, et al. 2010. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 363(9):809–19.
  • Fu C, Ma T, Zhou L, Mi QS, Jiang A. 2022. Dendritic cell-based vaccines against cancer: challenges, advances and future opportunities. Immunol Invest. 1–26. doi:10.1080/08820139.2022.2109486
  • Ghoneim HE, Fan Y, Moustaki A, Abdelsamed HA, Dash P, Dogra P, Carter R, Awad W, Neale G, Thomas PG, et al. 2017. De Novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell. 170(1):142–57 e19.
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. 2010. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 363(5):411–22.
  • Knight DA, Ngiow SF, Li M, Parmenter T, Mok S, Cass A, Haynes NM, Kinross K, Yagita H, Koya RC, et al. 2013. Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J Clin Invest. 123(3):1371–81.
  • Koguchi Y, Redmond WL. 2022. A novel class of on-treatment cancer immunotherapy biomarker: trough levels of antibody therapeutics in peripheral blood. Immunol Invest. 1–17. doi:10.1080/08820139.2022.2131570
  • Lim WA, June CH. 2017. The principles of engineering immune cells to treat cancer. Cell. 168(4):724–40.
  • Long M, Mims AS, Li Z. 2022. Factors affecting the cancer immunotherapeutic efficacy of T cell bispecific antibodies and strategies for improvement. Immunol Invest. 1–39. doi:10.1080/08820139.2022.2131569
  • Lurquin C, Van Pel A, Mariame B, De Plaen E, Szikora JP, Janssens C, Reddehase MJ, Lejeune J, Boon T. 1989. Structure of the gene of tum- transplantation antigen P91A: the mutated exon encodes a peptide recognized with Ld by cytolytic T cells. Cell. 58(2):293–303.
  • Mellman I, Turley SJ, Steinman RM. 1998. Antigen processing for amateurs and professionals. Trends Cell Biol. 8(6):231–37.
  • Mihalyo MA, Hagymasi AT, Slaiby AM, Nevius EE, Adler AJ. 2007. Dendritic cells program non-immunogenic prostate-specific T cell responses beginning at early stages of prostate tumorigenesis. Prostate. 67(5):67536–46.
  • Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, Yates KB, Lako A, Felt K, Naik GS, et al. 2019. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 20(3):326–36.
  • Milstein C. 1999. The hybridoma revolution: an offshoot of basic research. Bioessays. 21(11):966–73.
  • Palucka AK, Ueno H, Fay JW, Banchereau J. 2007. Taming cancer by inducing immunity via dendritic cells. Immunol Rev. 220(1):220129–50.
  • Ryder LP, Svejgaard A, Dausset J. 1981. Genetics of HLA disease association. Annu Rev Genet. 15(1):15169–87.
  • Scarlett UK, Rutkowski MR, Rauwerdink AM, Fields J, Escovar-Fadul X, Baird J, Cubillos-Ruiz JR, Jacobs AC, Gonzalez JL, Weaver J, et al. 2012. Ovarian cancer progression is controlled by phenotypic changes in dendritic cells. J Exp Med. 209(3):495–506.
  • Scheper W, Kelderman S, Fanchi LF, Linnemann C, Bendle G, de Rooij MAJ, Hirt C, Mezzadra R, Slagter M, Dijkstra K, et al. 2019. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med. 25(1):89–94.
  • Sharabi AB, Lim M, DeWeese TL, Drake CG. 2015. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 16(13):e498–509.
  • Sharma P, Allison JP. 2015. The future of immune checkpoint therapy. Science. 348(6230):56–61.
  • Simoni Y, Becht E, Fehlings M, Loh CY, Koo SL, Teng KWW, Yeong JPS, Nahar R, Zhang T, Kared H, et al. 2018. Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature. 557(7706):575–79.
  • Snell GD. 1981. Studies in histocompatibility. Science. 213(4504):172–78.
  • Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L, Castillo Gutierrez E, Rutkowski P, Gogas HJ, Lao CD, De Menezes JJ, et al. 2022. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 386(1):24–34.
  • Tran E, Robbins PF, Rosenberg SA. 2017. ‘Final common pathway’ of human cancer immunotherapy: targeting random somatic mutations. Nat Immunol. 18(3):255–62.
  • Ward Grados DF, Ahmadi H, Griffith TS, Warlick CA. 2022. Immunotherapy for bladder cancer: latest advances and ongoing clinical trials. Immunol Invest. 1–26. doi:10.1080/08820139.2022.2118606
  • Wherry EJ, Kurachi M. 2015. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 15(8):486–99.
  • Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, McNamara KL, Granja JM, Sarin KY, Brown RA, et al. 2019. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 25(8):1251–59.
  • Zinkernagel RM, Doherty PC. 1974. Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature. 248(5450):701–02.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.