Clinical Oncology: Case Reports

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.

Mini Review, Clin Oncol Case Rep Vol: 5 Issue: 9

An Examination of Cancer Immunotherapy: Past, Present, and Future

Elena Johnson*

Editorial Office, Clinical Oncology: Case Reports, United Kingdom

*Corresponding Author: Elena Johnson
Editorial Office, Clinical Oncology: Case Reports, United Kingdom
E-mail: oncologyreport@escienceopen.com

Received: September 05, 2022; Manuscript No: COCR-22-76684;
Editor Assigned: September 07, 2022; PreQC Id: COCR-22-76684 (PQ);
Reviewed: September 21, 2022; QC No: COCR-22-76684 (Q);
Revised: September 23, 2022; Manuscript No: COCR-22-76684 (R);
Published: September 30, 2022; DOI: 10.4172/cocr.5(9).253

Citation: Johnson E (2022) An Examination of Cancer Immunotherapy: Past, Present, and Future. Clin Oncol Case Rep 5:9

Abstract

Cancer immunotherapy has significantly increased patients' chances of survival and quality of life as compared to earlier standards of care (such as chemotherapy, radiation, and surgery). From the metastatic stage to the adjuvant and neoadjuvant settings in many cancer types, immunotherapy has now firmly established itself as a novel pillar of cancer care. In this review article, we emphasize how the development of cancer immunotherapy led to findings that are today considered best practices. We also discuss the existing drawbacks and restrictions of cancer checkpoint immunotherapy and the emerging approaches being used to address these problems in the fields of tailored cancer vaccines, autoimmunity, the microbiome, the tumor microenvironment, and metabolomics.

Keywords: Metabolomics; Personalized cancer vaccines; Immune-related adverse events; Immune checkpoint inhibitors

Introduction

The treatment of cancer patients has changed significantly thanks to the field of immuno-oncology. In the late 19th century, William B. Coley, who is now commonly regarded as the father of immunotherapy, made the first attempts to use the immune system's combative capabilities to cure cancer. He observed that some patients with substantial postoperative wound infections, which were frequent when aseptic technique had not yet been refined, would experience spontaneous regression of their unresected tumors while undergoing surgery on patients with bone sarcomas [1]. In an effort to cause sepsis and potent immunological and antitumor responses, Coley began injecting mixes of live and inactivated bacteria, including Serratia marcescens and Streptococcus pyogenes, into more than a thousand patients in 1891.

Before Coley's toxin's methods of action could be better understood in relation to the major mediators of sepsis, it would be more than fifty years. These mediators belong to the cytokine family, which also includes chemokines, interleukins, and interferons. The race to incorporate such cutting-edge discoveries into cancer therapy has begun once more [2]. With this unique strategy, doctors and researchers had only little success, occasionally producing clinical remissions in metastatic renal cell carcinoma with highdose interleukin 2 (il-2) and questionable responses in stages 3 and 4 melanoma with interferon. These modest successes were frequently offset by serious negative incidents. Only a tiny, carefully chosen minority of cancer patients would benefit from these innovative delivery techniques, such as pegylation, due to the unpredictable and sporadic immunological reactions they elicited in patients [3].

With a deeper understanding of the method by which innate immune cells destroy cancer cells-immune surveillance-the field of cancer immunotherapy experienced its next revolutionary wave. The key job of those hardwired signals is to keep the delicate balance between autoimmunity and immune surveillance against invading infections or aberrant cells. The heightened autoimmunity caused by blocking certain T cell surface receptors provides an immune response against tumors but also raises the risk of autoimmune responses [3].

Overview of checkpoint inhibitors

Cancer immuno-editing is the process by which different immune system components safeguard the host against the establishment of primary tumors or promote tumor escape, or both, by either sculpting tumor immunogenicity or attenuating antitumor immune responses. Immune checkpoints, which are immunological-cell surface receptors that regulate either the activation or suppression of immune responses, strictly regulate the process. On the one hand, activating the immune system is what is needed to prevent tumor growth, but it is also what causes autoimmunity. By upregulating immune activation at different stages of the immunological cycle, the discovery and development of monoclonal antibodies against the inhibitory immune checkpoints ctla-4 and PD-1 have produced remarkable antitumor responses. Therapies using immune checkpoint inhibitors are increasingly often prescribed for a variety of cancer types. Numerous clinical trials that are still in progress also examine how additional agonistic or inhibitory checkpoints may influence tumor-related results. The potential of the checkpoints varies [4]. In contrast to the CD28 antibody, which even at extremely low dosages caused a large cytokine crisis and the intensive care hospitalization of the first six healthy volunteers treated8, the agonistic OX40 antibody shows moderate clinical activity. In light of this, clinical research is still ongoing to determine the best treatment combination to cause the ideal level of immunological activation.

Modulating and predicting immune toxicity for better efficacy

Immune-Related Adverse Events (iraes), an immune activation and inflammatory reaction to the host's healthy tissues, are frequently immunotherapies' main drawbacks. The intended result is immune activation against the host's tumor, but iraes are difficult to forecast, identify, and cure. The addition of a ctla-4 antibody to PD-1 treatment in the context of metastatic melanoma is linked with just a little improvement in survival, but at the expense of more than doubling the rate of major adverse events. One patient in every 77 who received treatment with a combination was reported to die, according to a recent meta-analysis. For some illnesses, such immune-related myocarditis, the mortality rate among treated patients might reach 50%. General immunosuppression with corticosteroids, followed by one or more biologics (tumor necrosis factor inhibitors) or T cell suppressants is advised for serious iraes, according to recommendations (such as mycophenolate mofetil) [5]. The effects of those medicines on outcomes connected to cancer have not been fully elucidated in the future. An examination of baseline corticosteroid use in lung cancer patients revealed a correlation with poorer survival results. The administration of high-dose steroids in individuals with metastatic melanoma who had immune-related hypophyses was similarly linked to a worse prognosis. Contrarily, neither a diminished response to ici treatment nor survival was linked to the use of corticosteroids in other therapeutic contexts when patients had iraes.

In order to prevent reducing the effectiveness of Immune check point inhibitors, more research is required to determine the ideal immunosuppressive regimen to be utilized with them. In the specific context of organ transplantation, the use of mechanistic target of rapamycin inhibitors holds potential for reducing toxicities without decreasing efficacy [6].

A new era for tumor-specific vaccines in combination with Immune Checkpoint Inhibitor (ICIs)

Although ICIs has shown promising results, single-agent PD-1 inhibitors have an objective response rate that varies from almost nonexistent in pancreatic cancer and microsatellite-stable colonic adenocarcinoma to an average of 15%-30% in most other tumor types, but 50%-80% in melanoma, Hodgkin lymphoma, squamouscell carcinoma of the skin, and Merkel cell carcinoma. The response rate is increased with the addition of an anti-CTLA-4 drug, although the rate of toxicity is considerably increased. Combining an ICI with a therapy that can pre-sensitize the host's immune system to the tumor has been shown to be a sensible way to increase response rates without triggering autoimmunity. Tumor-specific vaccinations based on individualized neo-antigens exhibit great promise, according to recent investigations.

Solid tumors either lack this antigen or experience mutations under natural selection when exposed to therapeutic interventions such as monoclonal antibodies, unlike hematologic malignancies where a common antigen is uniformly expressed on the surface of all malignant cells making them amenable to targeted therapies such as therapy with chimeric antigen receptor T cells [7]. Traditional cancer vaccines have been ineffective for a variety of reasons, including as poor target antigen selection, a lack of immunogenicity, or poor patient selection. The effectiveness of cancer vaccines in the modern era depends on computational processes designed to quickly identify individual candidate neo-antigens. Whole-exome sequencing allows for thorough mutation analysis, and neo-epitopes generated by somatic mutations in the tumor are chosen based on affinity predictions because they are more likely to be recognized by the person's major histocompatibility class molecules. NetMHCpan (DTU Health Tech, Technical University of Denmark, Kongens Lyngby, Denmark), one of the most widely used prediction algorithms for major histocompatibility class I binding, relies on cutting-edge neural networks, highlighting the current power of bioinformatics for directing precision immuno-oncology.

Discussion

Cancer immunotherapy has significantly improved patients' chances of survival and quality of life. There are currently very few predictors of response and toxicity because not all tumors are created equal. Immuno-oncology is still in its relative infancy despite the quick advancements achieved in the field, and there are still many difficulties and obstacles to be overcome. The understanding that the traditional methods for evaluating treatment options in the age of chemotherapy and targeted therapies might not be appropriate for the new immunotherapies evolved over time. In order to account for the unexpected patterns of response observed during immunotherapy, such as cancer pseudo progression, the Response Evaluation Criteria in Solid Tumors (recist), which was used to evaluate response to treatments, was changed to develop irecist [8]. In the same way that TNM staging was essential in directing therapies during the chemotherapy era; new methods are needed during the cancer immunotherapy era. It has already been established that the Immuno-score enhances colon cancer TNM staging by providing crucial prognostic data. Given that T cells are already universally acknowledged as the primary mediators of antitumor effectiveness with conventional treatment, using the Immuno-score to help determine the best course of treatment for various cancer types is an appealing alternative [9]. That choice does not, however, rule out the use of potential extra characteristics that might reveal more information about the particulars of each situation.

Increasing the efficacy of combination medicines that are already well-established in clinical practise is getting harder. Combining ctla-4 and PD-1 inhibition has produced an extraordinary five-year overall survival rate above 50% in metastatic melanoma. The same combination has been linked to an intention-to-treat population overall survival rate of more than 60% at 3 years in metastatic renal cell cancer. Few unique combinations in the vast field of ongoing early-phase clinical trials have attained a level of efficacy comparable to those new standards of care. Their safety profiles most definitely need to be enhanced [10].

Conclusion

In conclusion, as described in the present review, the future of cancer immunotherapy may rely on combination therapies that combine checkpoint inhibitors-but not with other novel checkpoint inhibitors-with personalized cancer vaccines and novel targeted therapies that target the host microbiome, tumor glycosylation, and the tumor microenvironment. The current broad "shotgun" strategy, which exposes everyone within the permitted indications to ICIs, will be replaced by therapies that are specifically catered to the characteristics that make each cancer and host a unique coupling as a result of advancements in those domains.

References

  1. McCarthy EF (2006) The toxins of William B. Coley and the treatment of bone and soft-tissue sarcomas. Iowa Orthopaedic J 26: 154. [Google Scholar] [Cross Ref]
  2. Dinarello CA (2007) Historical insights into cytokines. Eur J Immunol 37: 34-45. [Google Scholar] [Cross Ref]
  3. Lee S, Margolin K (2011) Cytokines in cancer immunotherapy. Cancers 3: 3856-3893. [Google Scholar] [Cross Ref]
  4. O’Donnell JS, Teng MWL, Smyth MJ (2019) Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol 16: 151-167. [Google Scholar] [Cross Ref]
  5. Moslehi JJ, Salem JE, Sosman JA, Lebrun-Vignes B, Johnson DB (2018) Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet 391: 933. [Google Scholar] [Cross Ref]
  6. Liudahl SM, Coussens LM (2018) B Cells as biomarkers: Predicting immune checkpoint therapy adverse events. J Clin Invest 128: 577-579. [Google Scholar] [Cross Ref]
  7. Dubin K, Callahan MK, Ren B, Khanin R, Viale A, et al. (2016) Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Comm 7: 1-8. [Google Scholar] [Cross Ref]
  8. Arbour KC, Mezquita L, Long N, Rizvi H, Auclin E, et al. (2018) Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol 36: 2872-2878. [Google Scholar] [Cross Ref]
  9. Barnett R, Barta VS, Jhaveri KD (2017) Preserved renal-allograft function and the PD-1 pathway inhibitor nivolumab. N Engl J Med 376: 191-192. [Google Scholar] [Cross Ref]
  10. Esfahani K, Miller Jr WH (2017) Reversal of autoimmune toxicity and loss of tumor response by interleukin-17 blockade. New Eng J Med 376: 1989-1991. [Google Scholar] [Cross Ref]
international publisher, scitechnol, subscription journals, subscription, international, publisher, science

Track Your Manuscript

Awards Nomination