Vaccination for cancer: Myth or reality

Immunomedicine Pub Date : 2021-08-02 DOI:10.1002/imed.1026
David Avigan MD
{"title":"Vaccination for cancer: Myth or reality","authors":"David Avigan MD","doi":"10.1002/imed.1026","DOIUrl":null,"url":null,"abstract":"<p>May 19, 2021</p><p>Vaccination targeting infectious pathogens has profoundly impacted public health and has led to the eradication of diseases that have plagued human history. The development of therapeutic vaccines for cancer has been pursued over many years in effort to harness the selectivity and potency of the immune system with the promise of greater efficacy and safety than standard cytotoxic therapy. The unique potency of immune therapy was highlighted by the observation that allogeneic hematopoietic stem cell transplantation is uniquely curative for a subset of patients with hematologic malignancies due to the graft versus tumor effect mediated by alloreactive lymphocytes.<span><sup>1</sup></span> Vaccine platforms were developed in an effort to elicit tumor specific immune responses to target established malignancy and provide immunologic memory to prevent recurrence. Initial studies focused on the introduction of tumor associated antigens as peptides, proteins or whole tumor cells, or lysate most commonly in the setting of advanced disease.<span><sup>2</sup></span> While clinical trials demonstrated immunologic responses and anecdotal disease regression, therapeutic efficacy was not clearly seen and several randomized trials did not show benefit over standard therapy.<span><sup>3</sup></span> As such, vaccination was often viewed as an unrealized promise subsequently displaced by other therapeutic strategies.</p><p>The efficacy of cancer immunotherapy has been recently transformed with the enhanced understanding of the immunoregulatory aspects of the tumor microenvironment.<span><sup>4, 5</sup></span> The role of negative costimulatory signaling as a mediator of T cell exhaustion led to the development of checkpoint blockade as effective therapy for diverse malignancies, particularly when characterized by high mutational burden and the presence of tumor specific neoepitopes. In addition, CAR T cell therapy involving the ex vivo generation of effector cells with high levels of costimulatory molecule expression have received FDA approval for treatment of patients with lymphoma, acute lymphocytic leukemia, and multiple myeloma demonstrating a profound impact on a subset of patients with advanced disease. In this context, vaccine design has similarly evolved to incorporate this increased understanding of the complex interface between tumor cells and the immune environment to augment therapeutic efficacy, identify the optimal settings of intervention, and develop combinatorial approaches.</p><p>A critical factor for vaccine design is the identification of antigenic targets that are selectively expressed by malignant cells and potentially recognized by the T cell repertoire.<span><sup>6</sup></span> These have included aberrantly expressed oncogenic proteins, tissue specific markers, and antigens characteristically expressed in fetal development that are upregulated in the setting of malignancy.<span><sup>7</sup></span> While selection of antigen(s) is optimized when expression captures clonal diversity of the tumor, there has been growing appreciation of providing a platform of activation that facilitates epitope spreading to target cancer heterogeneity. In addition, efforts to enhance immunogenicity may involve generation of heteroclitic peptides that enhance T cell affinity.<span><sup>8</sup></span> Neoepitopes arising from mutational events may be identified through computational platforms that are recognized as foreign and targeted by high affinity T cells that have not been subjected to central or peripheral tolerance mechanisms to protect against auto-immunity.<span><sup>9, 10</sup></span></p><p>Another critical factor for enhancing vaccine efficacy is the effective presentation of antigen in the context of co-stimulation through recruitment of mediators of innate immunity and antigen presentation. Dendritic cells (DCs) express high levels of costimulatory molecules and inflammatory cytokines necessary for activation and expansion of primary tumor specific immunity.<span><sup>11</sup></span> DCs may be recruited in vivo through the use of oncolytic or immunogenic viral vectors or cytokines such as GM-CSF.<span><sup>12-15</sup></span> Alternatively, the ex vivo generation of antigen presenting cells provides a critical platform the generation of effective immunity. Loading of individual tumor antigens onto DCs may be accomplished through the use of tumor associated peptides, proteins, or corresponding RNA or DNA.<span><sup>16</sup></span> Alternatively, introduction of antigens derived from whole tumor cells have involved the use of tumor lysate, apoptotic bodies, allogeneic, and whole cell RNA. The relative immunologic potency of the different antigen platforms has not been fully elucidated but polyvalent vaccines may provide protection from tumor escape due to the emergence of antigen negative variants. We have developed hybridomas involving the fusion of patient derived tumor cells and autologous DCs capable of activating and expanding a diverse array of T cell clones targeting both shared and neoepitopes expressed by the malignant cell.<span><sup>16, 17</sup></span> Further enhancement of the vaccine platforms has been explored through the incorporation of checkpoint blockade or immunostimulatory platforms such as biomatrices facilitating antigen presentation.</p><p>The development of next generation vaccine platforms has led to the heightened expansion of tumor reactive T cell clones. Clinical trials have demonstrated an association between immunologic response and disease response or time to progression.<span><sup>7</sup></span> Of note, vaccination has been more recently largely pursued in the setting of low volume disease following cytoreduction to allow for the expansion of tumor reactive clones and minimize the immunoregulatory impact of a rapidly expanding tumor volume. Provocative results have been noted with respect to prolonged period of disease control but this has been difficult verify in large randomized clinical trials in which biologic diversity and a rapidly changing standard of care may also impact results.<span><sup>18</sup></span> Of note, in one example vaccination did not result in a statistically significant difference in time to progression but did appear to be associated with prolongation of survival raising a fundamental question as to how to best assess the impact of immunomodulatory therapy on long term outcome.</p><p>The increased understanding of the complex nature of immune regulation and cancer immunity has also suggested that combinatorial strategies might be the optimal approach to reverse tumor mediated immune suppression to create long term disease control. Effective vaccine platforms have demonstrated the capacity to elicit the expansion and transient activation of tumor reactive clones. However, functional potency of these clones may be limited by negative immune regulation of the tumor microenvironment. In contrast, immunostimulatory agents such as checkpoint inhibition may require the presence of tumor reactive lymphocytes as a substrate for therapeutic efficacy. As such, the combination of vaccination with agents targeting critical aspects of the tumor microenvironment may be required to achieve therapeutic efficacy.<span><sup>19, 20</sup></span> Similarly, immune effector cell therapy such as CAR T cells has shown short term potency in the setting of advanced disease where the antigenic target is not expressed on vital normal tissues. However, the hyper-stimulated nature of these cells may also result in disease escape due the induction of exhaustion and emergence of antigen negative variants. Vaccination may play a critical role in providing cyclic stimulation and facilitating epitope spreading for immune effector cells.<span><sup>21</sup></span></p><p>The development of therapeutic cancer vaccines has yet to realize to its considerable potential likely due to the barriers of overcoming tumor mediated immune suppression and tolerance mechanisms. A fundamental challenge involves the induction of clinically meaningful immunity that discriminates between tumor cells and normal tissue in which the patterns of antigen expression may significantly overlap. However, vaccine mediated stimulation may provide more durable expansion of tumor reactive clones utilizing central pathways of adaptive immunity that when combined with immunoregulatory agents and effector cell activation may provide sustained protection against malignancy.</p>","PeriodicalId":73348,"journal":{"name":"Immunomedicine","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2021-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/imed.1026","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Immunomedicine","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/imed.1026","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

May 19, 2021

Vaccination targeting infectious pathogens has profoundly impacted public health and has led to the eradication of diseases that have plagued human history. The development of therapeutic vaccines for cancer has been pursued over many years in effort to harness the selectivity and potency of the immune system with the promise of greater efficacy and safety than standard cytotoxic therapy. The unique potency of immune therapy was highlighted by the observation that allogeneic hematopoietic stem cell transplantation is uniquely curative for a subset of patients with hematologic malignancies due to the graft versus tumor effect mediated by alloreactive lymphocytes.1 Vaccine platforms were developed in an effort to elicit tumor specific immune responses to target established malignancy and provide immunologic memory to prevent recurrence. Initial studies focused on the introduction of tumor associated antigens as peptides, proteins or whole tumor cells, or lysate most commonly in the setting of advanced disease.2 While clinical trials demonstrated immunologic responses and anecdotal disease regression, therapeutic efficacy was not clearly seen and several randomized trials did not show benefit over standard therapy.3 As such, vaccination was often viewed as an unrealized promise subsequently displaced by other therapeutic strategies.

The efficacy of cancer immunotherapy has been recently transformed with the enhanced understanding of the immunoregulatory aspects of the tumor microenvironment.4, 5 The role of negative costimulatory signaling as a mediator of T cell exhaustion led to the development of checkpoint blockade as effective therapy for diverse malignancies, particularly when characterized by high mutational burden and the presence of tumor specific neoepitopes. In addition, CAR T cell therapy involving the ex vivo generation of effector cells with high levels of costimulatory molecule expression have received FDA approval for treatment of patients with lymphoma, acute lymphocytic leukemia, and multiple myeloma demonstrating a profound impact on a subset of patients with advanced disease. In this context, vaccine design has similarly evolved to incorporate this increased understanding of the complex interface between tumor cells and the immune environment to augment therapeutic efficacy, identify the optimal settings of intervention, and develop combinatorial approaches.

A critical factor for vaccine design is the identification of antigenic targets that are selectively expressed by malignant cells and potentially recognized by the T cell repertoire.6 These have included aberrantly expressed oncogenic proteins, tissue specific markers, and antigens characteristically expressed in fetal development that are upregulated in the setting of malignancy.7 While selection of antigen(s) is optimized when expression captures clonal diversity of the tumor, there has been growing appreciation of providing a platform of activation that facilitates epitope spreading to target cancer heterogeneity. In addition, efforts to enhance immunogenicity may involve generation of heteroclitic peptides that enhance T cell affinity.8 Neoepitopes arising from mutational events may be identified through computational platforms that are recognized as foreign and targeted by high affinity T cells that have not been subjected to central or peripheral tolerance mechanisms to protect against auto-immunity.9, 10

Another critical factor for enhancing vaccine efficacy is the effective presentation of antigen in the context of co-stimulation through recruitment of mediators of innate immunity and antigen presentation. Dendritic cells (DCs) express high levels of costimulatory molecules and inflammatory cytokines necessary for activation and expansion of primary tumor specific immunity.11 DCs may be recruited in vivo through the use of oncolytic or immunogenic viral vectors or cytokines such as GM-CSF.12-15 Alternatively, the ex vivo generation of antigen presenting cells provides a critical platform the generation of effective immunity. Loading of individual tumor antigens onto DCs may be accomplished through the use of tumor associated peptides, proteins, or corresponding RNA or DNA.16 Alternatively, introduction of antigens derived from whole tumor cells have involved the use of tumor lysate, apoptotic bodies, allogeneic, and whole cell RNA. The relative immunologic potency of the different antigen platforms has not been fully elucidated but polyvalent vaccines may provide protection from tumor escape due to the emergence of antigen negative variants. We have developed hybridomas involving the fusion of patient derived tumor cells and autologous DCs capable of activating and expanding a diverse array of T cell clones targeting both shared and neoepitopes expressed by the malignant cell.16, 17 Further enhancement of the vaccine platforms has been explored through the incorporation of checkpoint blockade or immunostimulatory platforms such as biomatrices facilitating antigen presentation.

The development of next generation vaccine platforms has led to the heightened expansion of tumor reactive T cell clones. Clinical trials have demonstrated an association between immunologic response and disease response or time to progression.7 Of note, vaccination has been more recently largely pursued in the setting of low volume disease following cytoreduction to allow for the expansion of tumor reactive clones and minimize the immunoregulatory impact of a rapidly expanding tumor volume. Provocative results have been noted with respect to prolonged period of disease control but this has been difficult verify in large randomized clinical trials in which biologic diversity and a rapidly changing standard of care may also impact results.18 Of note, in one example vaccination did not result in a statistically significant difference in time to progression but did appear to be associated with prolongation of survival raising a fundamental question as to how to best assess the impact of immunomodulatory therapy on long term outcome.

The increased understanding of the complex nature of immune regulation and cancer immunity has also suggested that combinatorial strategies might be the optimal approach to reverse tumor mediated immune suppression to create long term disease control. Effective vaccine platforms have demonstrated the capacity to elicit the expansion and transient activation of tumor reactive clones. However, functional potency of these clones may be limited by negative immune regulation of the tumor microenvironment. In contrast, immunostimulatory agents such as checkpoint inhibition may require the presence of tumor reactive lymphocytes as a substrate for therapeutic efficacy. As such, the combination of vaccination with agents targeting critical aspects of the tumor microenvironment may be required to achieve therapeutic efficacy.19, 20 Similarly, immune effector cell therapy such as CAR T cells has shown short term potency in the setting of advanced disease where the antigenic target is not expressed on vital normal tissues. However, the hyper-stimulated nature of these cells may also result in disease escape due the induction of exhaustion and emergence of antigen negative variants. Vaccination may play a critical role in providing cyclic stimulation and facilitating epitope spreading for immune effector cells.21

The development of therapeutic cancer vaccines has yet to realize to its considerable potential likely due to the barriers of overcoming tumor mediated immune suppression and tolerance mechanisms. A fundamental challenge involves the induction of clinically meaningful immunity that discriminates between tumor cells and normal tissue in which the patterns of antigen expression may significantly overlap. However, vaccine mediated stimulation may provide more durable expansion of tumor reactive clones utilizing central pathways of adaptive immunity that when combined with immunoregulatory agents and effector cell activation may provide sustained protection against malignancy.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
癌症疫苗接种:神话还是现实
16,17通过结合检查点阻断或免疫刺激平台(如促进抗原呈递的生物基质),探索了进一步增强疫苗平台的方法。下一代疫苗平台的发展导致肿瘤反应性T细胞克隆的高度扩张。临床试验已经证明免疫反应与疾病反应或进展时间之间存在关联值得注意的是,最近主要是在细胞减少后的小体积疾病中进行疫苗接种,以允许肿瘤反应性克隆的扩大,并最大限度地减少肿瘤体积迅速扩大对免疫调节的影响。18 .在长期疾病控制方面,已注意到令人振奋的结果,但这很难在大型随机临床试验中得到证实,在这些试验中,生物多样性和迅速变化的护理标准也可能影响结果值得注意的是,在一个例子中,接种疫苗并没有导致进展时间的统计学显著差异,但似乎确实与生存期延长有关,这就提出了一个基本问题,即如何最好地评估免疫调节治疗对长期结果的影响。对免疫调节和癌症免疫的复杂性的进一步了解也表明,组合策略可能是逆转肿瘤介导的免疫抑制以实现长期疾病控制的最佳方法。有效的疫苗平台已经证明能够引起肿瘤反应性克隆的扩增和短暂激活。然而,这些克隆的功能效力可能受到肿瘤微环境负性免疫调节的限制。相反,免疫刺激剂如检查点抑制可能需要肿瘤反应性淋巴细胞的存在作为治疗效果的底物。因此,可能需要将疫苗接种与靶向肿瘤微环境关键方面的药物结合起来才能达到治疗效果。19,20同样,免疫效应细胞疗法,如CAR - T细胞,在晚期疾病的情况下,抗原靶点不在重要的正常组织上表达,显示出短期效力。然而,这些细胞的过度刺激性质也可能导致由于诱导衰竭和抗原阴性变异的出现而导致疾病逃逸。疫苗接种可能在为免疫效应细胞提供循环刺激和促进表位扩散方面发挥关键作用。21 .治疗性癌症疫苗的开发尚未发挥其巨大潜力,这可能是由于克服肿瘤介导的免疫抑制和耐受机制的障碍。一个基本的挑战涉及诱导临床有意义的免疫,区分肿瘤细胞和正常组织,其中抗原表达模式可能显著重叠。然而,疫苗介导的刺激可能利用适应性免疫的中心途径提供更持久的肿瘤反应性克隆扩增,当与免疫调节剂和效应细胞激活相结合时,可能提供对恶性肿瘤的持续保护。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Issue Information Engineered immunologic niche monitors checkpoint blockade response and probes mechanisms of resistance The use of transcriptomic data in developing biomarkers in breast cancer Issue Information Purinergic signaling and purine base metabolism at the crossroads between immunity, metabolism, and cancer: A review
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1