Revolutionizing Cancer Treatment: A Guide to Immunotherapies
Renato Brito Baleeiro, Ph.D
Principal Scientist | Immuno-oncology | Cancer Vaccines and Immunotherapy | Drug Discovery | Project Leader | R&D
In recent years, the landscape of cancer treatment has undergone a remarkable transformation with the advent of immunotherapies. This groundbreaking approach harnesses the body's own immune system to combat cancer cells, marking a paradigm shift in how we perceive and address the complexities of this relentless disease.
?
Understanding Immunotherapy: A Game-Changer in Cancer Care
?Immunotherapy works by stimulating the immune system to recognize and attack cancer cells. Unlike traditional treatments such as chemotherapy and radiation, which directly target cancer cells, immunotherapies empower the body to fight off the disease naturally. This not only minimizes the side effects often associated with conventional treatments but also holds the promise of more effective and long-lasting results.
?
Immunostimulants: Provoking the Immune Response
?Immunostimulants serve as catalysts for the immune system by inducing responses through various mechanisms. These mechanisms include the production of cytokines, the release of interferons, and the activation of lymphocytes. The historical groundwork laid by observations such as Coley's in the early twentieth century and subsequent advancements with Bacillus Calmette–Guerin (BCG) and interferon-α (IFN-α) paved the way for immunostimulant applications in cancer management. Despite early promise, the acceptance of some immunostimulants faced obstacles, primarily due to inconsistent results and inadequately designed studies.
?
Cancer Vaccination: Tailoring Defenses Against Tumors
?Cancer vaccination represents an approach where anti-tumor antigens in the form of peptides, proteins, mRNA or DNA are employed to stimulate T-cells against cancer. Tumor antigens, whether generated by somatic mutations (neoantigens) or non-mutated abnormal proteins, trigger immune responses. These responses present as major histocompatibility complex (MHC) class I molecules at the surface of tumor cells, inciting CD8+ T cells. Additionally, MHC class II fragments, presented by professional antigen-presenting cells (APC), are recognized by CD4+ T cells.
?However, the clinical significance of tumor antigens lies in their specificity for particular MHC molecules, limiting their applicability to those expressing the specific MHC molecule. Tumor antigens that can generate peptides engaging both MHC class I and II molecules lay the foundation for the development of peptide- and protein-based vaccines. Numerous vaccines have been synthesized and tested for effectiveness in different cancers. Early-generation vaccines demonstrated limited clinical significance, particularly in advanced cancer stages. Insights gained from these initial attempts provided valuable understanding of tumor cell reactivity to immunization effects.
?To enhance specificity and efficacy, modifications to peptide and protein-based vaccines have been proposed. Immunological adjuvants, such as aluminum salts, oil-in-water emulsions (MF59), nontoxic derivatives from Salmonella (MPL), and water-in-oil emulsions (Montanide ISA 51 and ISA 720), aim to facilitate gradual antigen release and subsequent amplification of immune responses.
?Another proposed modification involves the insertion of toll-like receptor ligands (TLRL) like TLR3L, TLR4L, TLR7/8L (imiquimod, resiquimod), and TLR9L (CpG). TLRLs activate APCs, with some exhibiting stimulatory potentials for both APCs and natural killer (NK) cells, initiating tumor cell death. Notably, TLR9L has shown effectiveness in stimulating the induction of tumor antigen-specified CD8+ T cells in advanced cancer patients.
?In 1986, gp96, an endoplasmic reticulum-residing member of HSP90 (heat shock proteins), was isolated from fibrosarcomas of mice after stimulation with methylcholanthrene A. It was found to function as a tumor rejection antigen. Extracellular HSP has been observed to play a stimulatory role for the immune system against tumorous tissue either by displaying immunogenic peptides originating from tumors or integrating innate and adaptive immunity through the secretion of chemokines, cytokines, and nitric oxide. Clinical applications of gp96 and HSP70 peptide-based vaccines extracted from autologous tumor lysate have been explored in late-stage melanoma, metastatic colorectal cancer, glioma, and renal cell carcinoma patients.
?While the HSP–peptide complex treatment incited an immune response in the majority of patients, the response remained confined to specific patient subgroups. Clinical trials involving brain tumor patients treated with recombinant HSP70 in an intra-tumoral manner after surgery exhibited complete clinical responses along with a buildup of Th1 cell-mediated immune responses and a decline in immunosuppressive Treg cell populations.
Personalized neoantigen-based vaccines
The landscape of therapeutic cancer vaccines has seen remarkable progress in the last decade, thanks to advancements in rapid and cost-effective sequencing technologies and the development of sophisticated neoantigen prediction algorithms. These innovations have paved the way for the development of personalized cancer vaccines. In 2017, groundbreaking studies demonstrated the effectiveness of tailored personalized vaccines in melanoma, marking a significant milestone. Recent strides in mRNA vaccine trials for pancreatic cancer have shown promising clinical outcomes, adding to the optimism. Notably, a shared mutation, KRAS, prevalent in approximately 25% of all cancers, has gained traction. This year's clinical trial focusing on patients with pancreatic and colorectal cancers with mutated KRAS has yielded positive results, highlighting the potential of personalized immunotherapy. These advancements underscore a promising future in cancer treatment, instilling hope for patients.
Monoclonal Antibodies and checkpoint inhibitors: Precision Targeting of Immunomodulation
?Monoclonal antibodies (mAbs) play a pivotal role in immunotherapeutic strategies aimed at modulating the immunosuppressive influence of cancer cells. The development of mAbs gained prominence with K?hler and Milstein's demonstration in 1975 of high-quantity mAbs displaying identical antigens. Various mAbs with diverse mechanisms of action have since been developed, including those opposing neoplastic activity, neutralizing trophic signaling, or stimulating the immune system against tumor cells.
?The initial therapeutic role investigations of mAbs faced challenges, possibly due to the incompatibility of mouse-derived mAbs with human immune effector molecules. However, the subsequent development of chimeric or fully humanized mAbs significantly improved their efficacy. Studies validating their effectiveness in hematological and solid tumors marked a shift towards personalized and effective therapeutic interventions. Examples include trastuzumab, bevacizumab, cetuximab, rituximab, panitumumab, and others.
?Immune checkpoint proteins, such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed death (PD-1), have emerged as critical regulators of T cell activation, balancing pro-inflammatory and anti-inflammatory signaling. PD-1 has two ligands, PD-L1 and PD-L2, with PD-L1 expressed on both tumor and immune cells. When coupled with PD-1, it inhibits T cell multiplication and cytotoxicity. Blocking these inhibitors with their antibodies resulted in satisfactory outcomes in in-vivo studies.
?FDA-approved mAb drugs since 2010 include Ipilimumab (anti-CTLA-4), Nivolumab, Pembrolizumab, and Cemiplimab (anti-PD-1), as well as Atezolimumab, Durvalumab, and Avelumab (anti PD-L). Ipilimumab and tremelimumab are mAbs formulated to counteract the activity of CTLA-4, thus allowing prolonged activation of T cells and enhancing T-cell mediated immunity along with the patient's anti-tumor immune response. Clinical trial data indicate low efficacy and high toxicity in patients treated with anti-CTLA-4 and resistance development in those treated with anti-PD-1 therapy.
领英推荐
?Ibritumomab and tositumomab radioconjugates deliver radioactive isotopes to intended cells. Alemtuzumab, binding to CD52 and leading to cellular lysis, is recommended by the FDA for fludarabine-refractory chronic lymphocytic leukemia (CLL), with reported clinical significance for cutaneous T-cell lymphoma, peripheral T-cell lymphoma, and T-cell prolymphocytic leukemia.
?With increased response rates and disease-free survival compared to chemotherapy, milder side effects generally caused by an allergic reaction due to the introduction of foreign proteins are observed. However, infrequent acute adverse effects such as arterial thromboembolic events in patients treated with Bevacizumab and autoimmune colitis caused by CTLA4 specific mAbs ipilimumab and tremelimumab have been observed.
?
Dendritic Cell Induction: Orchestrating Immune Responses
?Dendritic cells play a pivotal role in mediating innate immunity and stimulating adaptive immunity. Dysfunction of endogenous dendritic cells in cancer patients has led to the development of ex-vivo dendritic cells with controlled loading of antigens, enhancing the specificity and magnitude of the T-cell response. Ex-vivo generation allows the incorporation of supplementary features, such as tumor-relevant homing signals that direct the trafficking of immune cells toward potential metastatic sites.
?In-vivo dendritic cells have the potential to acquire resistance to inhibitory factors like IL-10, TGF-β, VEGF, and IL-6. However, an increase in regulatory T cells (Tregs) is observed in response to cancer vaccines, compromising the effectiveness of the vaccine.
?Clinical use of partially mature "first-generation" dendritic cells has been explored following successes in melanoma and follicular lymphoma. However, the expression of costimulatory molecules and immunogens remained below the optimal level compared to mature "second-generation" dendritic cells. To address this, an improvement in the macrophage-conditioned medium and in the cytokine cocktail, including IL-1α, tumor necrosis factor-α (TNF-α), IL-6, and PGE2, was introduced to stimulate dendritic cells and promote high expression of costimulatory molecules. Compared to immature dendritic cells, this cocktail exhibited enhanced immunogenic function along with an upgraded migratory response to lymph nodes.
?
Genetic Immunization of Cancers: Navigating the Challenges
?Various strategies have been proposed for genetically immunizing solid tumors, including cytokine gene therapy and plasmid-based immunization. Previous attempts, such as injecting plasmid DNA encoding cytokines to stimulate an immune response against tumor cells, faced challenges due to a limited immunogenic response. The plasmid-based immunization process, delivering antigens through viral and microbial vectors, has shown promising outcomes by eliciting both antibodies and cellular responses in mice.
?Clinical trials evaluating the effectiveness of self-tumor antigens (TAs), such as carcinoembryonic antigen (CEA) against colorectal cancer, confirmed the safety of DNA immunization. However, responsiveness to CEA varied among patients, highlighting the inadequacy of plasmid DNA immunization in stimulating a T-cell response. Similarly, a study involving MART-1 plasmid injection intramuscularly in melanoma patients reported no increase in immunity. The challenge lies in achieving a balanced response between neutralizing antibodies and the expanding population of Treg cells to self-antigens, hindering the improvement of immunity against cancer.
?A study utilizing an alpha-virus plasmid carrying the CEA antigen gene enveloped in virus-like replicon particles (VRP) claimed a reduction in the neutralization effect caused by antibodies and Treg cells, leading to an improvement in immunotherapeutic treatment and overall survival rate.
??
Chimeric Antigen Receptor Therapy (CAR-T): Unleashing Precision in Cancer Eradication
?Chimeric antigen receptor T (CAR-T) cell therapy stands as a beacon of promise due to its durable and effective complete responses. Engineered synthetic receptors guide lymphocytes, typically T cells, to recognize and eradicate cells expressing specific antigens. Since 2017, various CAR-T products have received FDA approval, targeting the CD19 antigen in large B-cell lymphoma.
?However, challenges such as antigen escape, observed in patients treated with single antigen-targeting CAR-T, result in the complete or partial loss of that antigen in a significant portion of relapsed/recurrence cases. Target antigens for multiple myelomas, such as B cell maturation antigen (BCMA) and CD38, have shown promise. Clinical trials for various solid malignancies, including glioblastoma, renal cell carcinoma, lung cancer, and hepatocellular carcinoma, are ongoing.
?Second, third, and fourth-generation CAR-T cells, with continual refinement and enhancement, signify progressive advancements in the design and functionality of CAR-T cells. These iterations represent a dynamic field of research and development, continually pushing the boundaries of CAR-T cell therapy for improved outcomes in cancer treatment.
?
Overcoming Challenges in Immunotherapy
?While immunotherapies offer unprecedented promise, challenges persist. Resistance mechanisms and potential side effects demand ongoing research and development. Scientists are actively exploring combination therapies and novel approaches to address these challenges, ensuring the continued evolution of immunotherapeutic strategies.
?
The Future of Cancer Treatment
?The evolution of immunotherapies marks a pivotal moment in the history of cancer treatment. As research progresses and technology advances, we anticipate even greater breakthroughs. The ongoing commitment to innovation and the relentless pursuit of understanding the complexities of the immune system hold the promise of transforming cancer into a manageable and, in some cases, curable disease.
Embracing your journey is essential to finding fulfillment in life. As Ralph Waldo Emerson once said, life is a journey, not a destination ~ Stay inspired, and keep seeking the beauty in every step of your journey. ??? #EternalLife #Journey #Inspiration
Medical Doctor/ MSc/ Immunologist/ Immunotherapy/ Advanced Therapies/ MSL/ Clinical Development/ Medical devices/ Regulatory Affairs/ The content reflects my professional opinion and not that of others
1 年Renato Brito Baleeiro, Ph.D excellent post. Thank you for sharing Easy IMMUNO-ATMPs