Promises and Challenges of TCR-engineered T-cell Therapy for Cancer
https://www.ucir.org/therapies/therapy-cell-therapy

Promises and Challenges of TCR-engineered T-cell Therapy for Cancer

In the last decade, the landscape of cancer treatment has been dramatically reshaped by immunotherapy. Within just seven years, the FDA has approved seven immune checkpoint inhibitors (ICIs) across more than 85 oncology indications, marking a significant stride forward in cancer therapy. However, despite these remarkable advancements, a notable proportion of patients fail to benefit from ICIs due to the limited presence of tumor-specific effector T cells.

Adoptive cell transfer (ACT), emerged as a promising approach offering renewed hope for cancer patients. ACT involves infusing antigen-specific T cells in quantities surpassing what the body can naturally produce—an innovation poised to overcome the aforementioned limitation. Among the array of ACT techniques under development, including tumor-infiltrating lymphocyte (TIL) therapy, T cell receptor–engineered T (TCR-T) cell therapy, and chimeric antigen receptor T (CAR-T) cell therapy, each holds distinct potential.

Originally, ACT focused on isolating tumor-specific TILs for ex vivo expansion and reintroduction into patients—an approach particularly effective in certain cancers like melanoma. However, its feasibility was limited to tumors amenable to resection with sufficient T cell availability for isolation and amplification. TCR-T and CAR-T cell therapies, on the other hand, present genetically engineered T cells expressing receptors targeting tumor antigens, showing substantial progress in hematological cancers.

While CAR-T therapies have shown remarkable success in hematological cancers—culminating in six FDA approvals targeting CD19 or B cell maturation antigen—their efficacy in solid tumors has faced challenges. These hurdles include antigen scarcity, tumor heterogeneity, and immunosuppression within the tumor microenvironment.

Amidst these challenges, TCR-T cell therapy emerges as a promising alternative, offering several advantages. Notably, TCR-T cells boast a broader repertoire of targetable antigens compared to CAR-T cells, owing to their ability to recognize epitopes presented by the major histocompatibility complex (MHC). Additionally, their heightened sensitivity and high avidity hold potential for improved tumor detection and killing.

Excitingly, compelling clinical data now support the efficacy of TCR-T cell therapies in solid cancers. This article aims to synthesize the diverse array of tumor antigens targeted by TCR-T cell therapy in clinical and preclinical settings. We will explore strategies for identifying tumor-specific TCRs, optimizing their expression, and enhancing the therapy's overall efficacy. Additionally, we'll delve into the challenges ahead, emphasizing the importance of addressing safety concerns while striving for improved outcomes.

?

Tumor Antigens Tested in Clinical Trials

In the realm of advancing TCR-T cell therapies, selecting the right antigens stands as a pivotal step toward crafting treatments that are both safe and effective. Picture the ideal antigen: one that is expressed solely and consistently within tumor cells, producing epitopes presented on MHC class I molecules on their surface. As clinical trials unfold, two major classes of tumor antigens are taking center stage: tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs).

TAAs, characterized by their overexpression in tumors compared to normal tissues, are split into two categories: tissue differentiation antigens (TDAs) and cancer germline antigens (CGAs). TDAs, such as MART-1 and gp100, have garnered attention due to their presence in tumor tissues. Yet, while TDAs offer promise as shared targets across patients, their expression in normal tissues poses challenges, potentially leading to on-target off-tumor toxicity. Clinical trials targeting TDAs like MART-1 and gp100 have shown some clinical responses, albeit with notable toxicities stemming from their low expression in normal tissues. Efforts to enhance clinical outcomes, such as using affinity-enhanced TCRs, have been met with mixed results, underscoring the complexity of balancing efficacy with safety in TCR-T cell therapy.

Meanwhile, CGAs, like members of the MAGE-A protein family and NY-ESO-1, have emerged as promising targets despite initial setbacks. Early trials targeting MAGE-A antigens encountered severe toxic effects due to cross-reactivity with normal tissues. However, recent advancements, including MHC class II–restricted TCRs and affinity-enhanced TCRs, have shown more encouraging results, particularly in sarcoma. Similarly, targeting NY-ESO-1 has shown promise in melanoma and synovial sarcoma, with substantial response rates and minimal toxicities observed in clinical trials.

On the frontier of tumor-specific antigens lie TSAs, or neoantigens, exclusively expressed by tumor cells. Targeting these neoantigens offers a distinct advantage: minimal risk of toxicity towards normal tissues. Mutation-associated neoantigens, derived from cancer-initiating genetic events, are paving the way for personalized TCR-T cell therapies. Clinical trials targeting mutations in genes like TP53 and KRAS have demonstrated efficacy with manageable toxicities, signaling a promising avenue for individualized treatment strategies. Moreover, the advent of CRISPR gene editing has opened new possibilities for personalized TCR-T cell therapies, offering a glimpse into the future of precision medicine.

Viral antigens, stemming from viral-induced cancers like HPV and HBV, present another avenue for TCR-T cell therapy. Clinical trials targeting viral antigens have shown notable response rates with minimal toxicities, showcasing their inherent tumor specificity. Additionally, alternative TSAs, derived from unconventional sources such as abnormal mRNA splicing and retroelements, are expanding the repertoire of targetable tumor antigens. While clinical data on these unconventional antigens are limited, their discovery holds promise for broadening the scope of TCR-T cell therapy.

?

Enhancing TCR Identification and Optimization

The quest for identifying epitope-specific T-cell receptors (TCRs) is a challenging yet rewarding endeavor, given the vast array of TCR repertoires and the intricacies of their interactions with peptide-major histocompatibility complex (pMHC) molecules. Fascinatingly, a single TCR can discern up to an impressive 10^6 different epitopes, while conversely, one epitope can be recognized by multiple TCRs. This intricate dance of recognition forms the basis of our understanding of immune responses and provides a solid foundation for innovative approaches in TCR identification and optimization.

The methodologies for pinpointing pMHC-specific TCRs typically revolve around in vitro induction of a T-cell response within a specific human leukocyte antigen (HLA) context. T cells, sourced directly from tumors or from the blood of patients or healthy donors, undergo antigen stimulation, or T cell priming, to enrich the population with antigen-specific T cells following clonal expansion. Subsequent sorting and in vitro amplification of epitope-specific T cells pave the way for TCR sequence determination, achieved through TCRα and TCRβ sequencing of isolated T cells, often employing rapid amplification of cDNA ends–polymerase chain reaction techniques. Recent advancements have integrated VDJ sequencing and reference sequences from the ImMunoGeneTics database, providing a more comprehensive approach to identifying functional TCRs. Additionally, the advent of single-cell RNA sequencing (scRNA-seq) has revolutionized TCR identification by focusing on TCR sequences (scTCR-seq), enabling the selection of highly functional TCRs early in the process. Combinations of innovative techniques, including barcoded tetramers, barcoded antibodies, and scRNA-seq, have further enhanced the identification of functional TCRs with peptide-specific activation signatures. These advancements pave the way for the simultaneous identification of TCRs specific for different epitopes, opening new avenues for research and therapeutic development.

Furthermore, recent progress in in vivo mouse models with humanized T cell repertoires has created unprecedented opportunities to identify human epitope-specific TCRs following in vivo priming or vaccination. These models offer advantages such as an increased likelihood of identifying high-affinity TCRs, particularly in scenarios where the absence of antigen expression prevented negative thymic selection. However, it's essential to note that while animal models offer valuable insights, TCR identification from human T cells remains pivotal to mitigate the risk of selecting self-reactive T cells, ensuring safety and efficacy in therapeutic applications.

Once the TCR sequence is elucidated, optimization becomes paramount to enhance TCR expression and/or affinity. For instance, TAA-specific TCRs may exhibit low affinity, necessitating methods to improve TCR affinity while balancing the potential pitfalls of overly strong interactions. Careful optimization aims to increase TCR affinity without compromising T cell functionality, considering the delicate balance required for effective tumor cell recognition and successive targeting.

High TCR expression and proper assembly are crucial for generating functional TCR-T cells. Recent studies emphasize the importance of global TCR conformation and interactions between variable and constant regions in ensuring optimal TCR expression. Strategic modifications, such as replacing suboptimal residues and engineering additional disulfide bonds, have shown promise in enhancing TCR stability and functionality. Moreover, advancements in genome editing technologies, particularly CRISPR-Cas9, offer innovative avenues for replacing endogenous TCRs with transgenic counterparts, minimizing mispairing issues and enhancing TCR-T cell potency.

Validation of the selected T cell engineering strategy is imperative to assess both efficacy and safety. Functional assays, including multimer binding assays and coculture experiments, provide insights into TCR assembly, specificity, and cytotoxicity. Moreover, in vivo studies utilizing engineered mouse models offer valuable data on TCR-T cell viability, functionality, and persistence, crucial parameters for evaluating therapeutic potential.

?

Addressing the Challenges and Opportunities in TCR-T Cell Therapies

TCR-T cell therapies have emerged as promising avenues in cancer treatment, offering a potent weapon against malignancies. While navigating challenges inherent in such therapies, significant strides have been made toward enhancing efficacy and safety, shaping a future brimming with hope and potential.

One critical aspect demanding attention is the prediction and management of toxicity associated with TCR-T cell therapies. In clinical settings, toxicities have been observed, primarily stemming from on-target off-tumor effects, where TCR-T cells target antigens expressed in normal tissues. However, these challenges are not insurmountable. Rigorous evaluation methods, including bioinformatic analysis, immunopeptidomics, and in vitro assays, enable us to anticipate and mitigate potential toxicities. Advances in technology, such as the development of CAR-T cells targeting specific pMHC complexes, open avenues for precise histological assessment, enhancing our ability to differentiate between tumor and normal tissues.

Equally significant are on-target off-tumor toxicities, often resulting from TCR recognition of unintended antigens on normal cells. Despite these hurdles, proactive strategies are evolving to assess TCR cross-reactivity at the preclinical stage. Techniques like alanine scanning and mutational positioning scans offer insights into potential cross-reactive epitopes, minimizing risks associated with affinity-enhanced TCR-T cells. Moreover, innovative approaches employing deep learning algorithms and structural modeling provide comprehensive frameworks for predicting and mitigating cross-reactivity, paving the way for safer and more effective therapies.

Amidst the quest for improved therapies, understanding resistance mechanisms is paramount. Primary resistance mechanisms, such as low antigen expression or T cell exhaustion, pose initial challenges. However, it is the secondary resistance mechanisms that demand close scrutiny. Upregulation of immune checkpoint ligands and loss of MHC class I expression on tumor cells represent formidable hurdles. Yet, recent findings underscore the dynamic nature of resistance mechanisms, offering insights into potential interventions. Strategies like epigenetic modulation and targeted therapies hold promise in overcoming resistance, heralding a new era in cancer immunotherapy.

As we continue to unravel the intricacies of TCR-T cell therapies, collaboration across disciplines and sustained investment in research are imperative. By embracing these challenges as opportunities for innovation, we chart a course towards transformative treatments, enriching the lives of patients and fostering optimism in the fight against cancer. Together, we stand poised at the threshold of a new dawn in cancer immunotherapy, guided by hope, resilience, and the relentless pursuit of progress.

?

Towards the Future of T-cell Therapy

Advancements in T cell engineering herald a promising era in the fight against solid tumors. While the path forward is illuminated by optimism, it's imperative to acknowledge the hurdles that demand our attention. T cell–mediated toxicities, resistance mechanisms, and accessibility stand as formidable challenges on this journey.

To mitigate the risks associated with T cell activation against normal cells, we explore innovative strategies inspired by logic gates, akin to those proposed for CAR-T cells. These gates, such as "A AND B" and "A NOT B," offer avenues to condition T cell activation or inhibition based on the integration of multiple signals. Though more intricate for TCRs compared to CARs, recent developments in inhibitory signaling platforms offer promising solutions.

Moreover, the deployment of suicide gene systems adds another layer of safety, serving as a fail-safe mechanism to prevent unforeseen adverse events. By equipping engineered T cells with a kill switch, we enhance control and bolster confidence in the therapeutic process.

Beyond the realm of toxicities, resistance mechanisms pose another formidable challenge. Yet, through strategic combinations, we pave the way for enhanced efficacy. Clinical trials marrying TCR-engineered T cells with immune checkpoint inhibitors (ICIs) showcase the potential of synergistic approaches. By targeting specific neoantigens and leveraging complementary therapies like pembrolizumab, we glimpse into a future where resistance is met with resilience.

Gene modifications borrowed from CAR-T cell therapies offer further avenues to subvert tumor-related immunosuppression. Disruption of PD-1, incorporation of PD-1–CD28 chimeric constructs, and manipulation of transforming growth factor–β receptor type 2 present promising strategies to enhance T cell function within the hostile tumor microenvironment.

A critical aspect of T cell therapy lies in the persistence of adoptively transferred T cells. By engineering T cells with a stem cell memory phenotype (Tscm), we extend their lifespan and bolster their antitumor efficacy. Through meticulous in vitro cultivation supplemented with interleukins, we nurture Tscm cells primed for enduring impact.

While current TCR-T cell therapies primarily rely on autologous T cell engineering, the prospect of allogeneic approaches holds immense potential. Swift availability, standardized production, and reduced costs underscore the appeal of allogeneic strategies. By leveraging technologies to circumvent graft-versus-host disease and immune rejection, we inch closer to realizing the promise of universal T cells.

In summation, the journey of TCR-T cell therapy is marked by significant strides and promising outcomes. Despite the inherent complexities, our resolve remains steadfast. With refined tumor-specific antigen selection and optimized T cell engineering, we navigate towards heightened efficacy and minimized toxicity.

Combining these efforts with complementary therapies promises to unlock the full potential of TCR-T cells. From enhancing T cell homing to fortifying their activity and persistence, synergistic approaches hold the key to transformative outcomes. Just as next-generation CAR-T cells have reshaped the landscape of immunotherapy, TCR-T cells stand poised to address unmet therapeutic needs and redefine the trajectory of cold tumors.

要查看或添加评论,请登录

社区洞察

其他会员也浏览了