1. Introduction
Cancer is a multifaceted disease involving uncontrolled cell proliferation and the ability to invade surrounding tissues and spread to distant organs. The vast heterogeneity among tumors, both within a single tumor and between patients, complicates effective treatment [
1,
2]. Despite advances in conventional therapies such as chemotherapy and radiation, these treatments often result in significant toxicity and poor outcomes due to tumor resistance mechanisms [
3]. As a result, recent efforts in cancer therapy have increasingly focused on more targeted approaches.
The advent of antibody-based therapies, precision medicine, and immunotherapies has revolutionized cancer treatment. The central goal of these approaches is to specifically target tumor cells while minimizing damage to healthy tissues. For instance, monoclonal antibodies (mAbs) have demonstrated significant efficacy in treating cancers like breast cancer (HER2-positive) and lymphoma [
4]. Furthermore, precision medicine, which tailors treatment based on genetic and molecular profiling, has opened new avenues for personalized care [
5]. In parallel, immunotherapies, such as immune checkpoint inhibitors and CAR-T cell therapies, have significantly improved patient outcomes by harnessing the body’s immune system to target and destroy cancer cells [
6].
Despite the impressive potential of these therapies, challenges remain, particularly with tumor heterogeneity, drug resistance, immune-related adverse events, and the accessibility of such treatments to the broader population. In this review, we explore the current advancements in cancer treatment, focusing on the impact of antibody-based therapies, precision medicine, and emerging immunotherapies, and we examine their clinical implications and future directions.
2. Antibody-Mediated Therapy
Background and Mechanisms
Monoclonal antibodies (mAbs) have transformed cancer therapy by providing highly targeted treatment options with lower toxicity compared to conventional chemotherapy. mAbs target specific antigens expressed on tumor cells and mediate anti-tumor effects through several mechanisms: (1) direct inhibition of tumor cell growth via receptor blocking (e.g., trastuzumab targeting HER2), (2) inducing immune-mediated destruction of tumor cells through antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) [
7,
8], and (3) serving as vehicles for targeted drug delivery in the form of antibody-drug conjugates (ADCs) [
9].
Monoclonal antibodies are often used in combination with other therapies, such as chemotherapy or radiation, to enhance therapeutic efficacy [
10]. For example, the combination of rituximab with chemotherapy has been highly effective in treating B-cell lymphomas and chronic lymphocytic leukemia (CLL) [
11].
The mechanisms by which monoclonal antibodies exert their effects in cancer therapy are summarized in
Table 1.
Table 1 outlines the primary mechanisms of action of monoclonal antibodies (mAbs) in cancer therapy, highlighting their specific targets and clinical applications. These mechanisms allow for highly targeted and effective treatment options compared to traditional chemotherapy.
Advances in Antibody Technology
Recent technological innovations have improved the efficacy and safety profiles of mAbs. Bispecific antibodies (BsAbs), such as blinatumomab, can engage both T-cells and tumor cells, thereby enhancing immune-mediated tumor elimination [
12]. ADCs, including trastuzumab emtansine and brentuximab vedotin, combine the specificity of mAbs with the cytotoxic power of chemotherapeutic agents, targeting tumors while reducing systemic toxicity [
13]. The clinical success of these therapies highlights their promise in treating difficult-to-target cancers.
Additionally, Chimeric Antigen Receptor T-cell (CAR-T) therapies represent one of the most significant advances in immunotherapy. CAR-T cells are autologous T-cells genetically modified to express receptors specific to tumor-associated antigens, enabling them to target and destroy malignant cells. Clinical trials have shown exceptional efficacy in hematologic malignancies, particularly in leukemia and lymphoma [
14]. However, the application of CAR-T therapies in solid tumors has been less successful due to challenges such as the immunosuppressive tumor microenvironment and antigen heterogeneity [
15].
Key advances in antibody-mediated cancer therapies, including bispecific antibodies and CAR-T cell therapies, are presented in
Table 2.
Table 2 summarizes some of the key advances in antibody-mediated therapies, including monoclonal antibodies, bispecific antibodies, and CAR-T cell therapies, along with their targets, mechanisms, and clinical applications. These treatments have significantly impacted the landscape of cancer therapy.
Clinical Successes and Challenges
While monoclonal antibodies and CAR-T cell therapies have shown significant promise, challenges remain. Tumor resistance to mAbs can arise through mechanisms such as antigen loss, antigen shedding, and altered tumor microenvironment [
16]. In CAR-T therapy, challenges like cytokine release syndrome (CRS) and neurotoxicity are significant hurdles [
17]. Moreover, the high cost and complex manufacturing processes associated with CAR-T therapies limit their accessibility and widespread use [
18]. Therefore, ongoing research is focused on optimizing CAR-T therapies and developing new-generation mAbs to address these limitations.
3. Precision Medicine and Genomic Profiling
Personalized Treatment Strategies
Precision medicine aims to tailor treatments based on an individual’s genetic and molecular profile, ensuring the most effective therapies are chosen for each patient. Next-generation sequencing (NGS) allows the identification of genetic mutations and alterations that drive tumor growth, enabling clinicians to select therapies that target specific genetic aberrations [
19]. For example, targeted therapies such as epidermal growth factor receptor (EGFR) inhibitors for non-small cell lung cancer (NSCLC) and BRAF inhibitors for melanoma have shown substantial clinical benefits [
20,
21].
Clinical Implementations
Precision oncology has transformed the management of cancers with known molecular drivers. Tyrosine kinase inhibitors (TKIs) such as imatinib for chronic myelogenous leukemia (CML) and crizotinib for ALK-positive NSCLC have been game-changers in cancer treatment [
22]. Despite the success of these targeted therapies, they are often limited by the development of secondary mutations or resistance, which necessitates the ongoing development of next-generation inhibitors to overcome these challenges [
23].
Precision medicine also relies on biomarker testing to predict treatment response. For instance, testing for microsatellite instability (MSI) or tumor mutational burden (TMB) can help identify patients who may benefit from immune checkpoint inhibitors, such as pembrolizumab or nivolumab [
24]. Additionally, liquid biopsy, which analyzes circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs), offers a non-invasive way to monitor disease progression and treatment response [
25].
Examples of targeted therapies in precision medicine and their clinical impact are provided in Table 3.
Table 3 highlights examples of targeted therapies used in precision medicine, demonstrating how these therapies are tailored to specific molecular alterations in tumors. Such treatments have led to significant clinical improvements in various cancers, including NSCLC and melanoma.
Emerging Biomarkers
Advancements in molecular profiling have led to the discovery of new biomarkers that predict response to therapy. In addition to traditional genetic mutations, new biomarkers such as TMB and MSI are increasingly used to guide treatment decisions, especially for patients with cancers that may be more responsive to immunotherapy [
26]. Liquid biopsy, which allows for dynamic monitoring of tumor evolution, holds the promise of improving patient management by detecting minimal residual disease and early signs of relapse [
27].
Emerging biomarkers, such as microsatellite instability (MSI) and tumor mutational burden (TMB), and their role in predicting response to therapies are detailed in
Table 4.
Table 4 presents emerging biomarkers, such as microsatellite instability (MSI) and tumor mutational burden (TMB), that are increasingly used to predict a patient's response to cancer therapies. Liquid biopsy and ctDNA are also helping clinicians monitor disease progression non-invasively.
4. Immunotherapy: Harnessing the Immune System
Checkpoint Inhibitors
Immune checkpoint inhibitors, including PD-1/PD-L1 inhibitors (e.g., pembrolizumab, nivolumab) and CTLA-4 inhibitors (e.g., ipilimumab), have revolutionized cancer treatment by blocking immune checkpoints that inhibit T-cell activity [
28]. By disrupting these checkpoint pathways, checkpoint inhibitors re-activate the immune system, enabling T-cells to attack tumor cells. These therapies have demonstrated remarkable clinical success in cancers such as melanoma, lung cancer, and bladder cancer [
29].
Recent studies have shown that combining immune checkpoint inhibitors with chemotherapy or targeted therapies may enhance treatment efficacy and overcome resistance mechanisms [
30]. For example, the combination of nivolumab and chemotherapy has shown promising results in patients with advanced NSCLC [
31]. However, immune-related adverse events (irAEs) remain a challenge, including autoimmune diseases and inflammation [
32].
CAR-T Cell Therapy
CAR-T cell therapies have demonstrated significant efficacy in hematologic cancers, particularly in acute lymphoblastic leukemia (ALL) and large B-cell lymphoma, where they can achieve remission rates of up to 80-90% [
33]. However, the application of CAR-T in solid tumors is still a subject of active research. Solid tumors present unique challenges such as antigen heterogeneity, an immunosuppressive tumor microenvironment, and physical barriers like the extracellular matrix [
34].
Oncolytic Viruses and Other Immunotherapies
Oncolytic viruses, which selectively infect and lyse cancer cells while stimulating immune responses, are emerging as promising cancer therapies. Studies have demonstrated their potential in combination with immune checkpoint inhibitors to enhance anti-tumor immunity [
35]. The combination of oncolytic virotherapy with other immunotherapies like CAR-T is also being explored in clinical trials [
36].
In addition to oncolytic viruses, bispecific T-cell engagers (BiTEs), such as blinatumomab, are designed to recruit T-cells to cancer cells and induce their destruction [
37]. These agents are showing promise in hematologic cancers and are being investigated for their potential in solid tumors.
The clinical successes and challenges in immunotherapy are summarized in
Table 5.
Table 5 provides an overview of the clinical successes and challenges associated with various immunotherapies, including immune checkpoint inhibitors, CAR-T cell therapy, and oncolytic viruses. While these therapies have revolutionized cancer treatment, challenges such as immune-related adverse events and therapy resistance remain.
5. Combination Therapies and Multi-Omics Approaches
Synergy in Treatment
Combining different therapeutic modalities, such as chemotherapy, targeted therapy, and immunotherapy, has shown promising results in overcoming resistance and improving patient outcomes. Combination strategies can take advantage of the synergistic effects of various treatments to attack tumors from multiple angles. For example, combining immune checkpoint inhibitors with chemotherapy has enhanced response rates in patients with metastatic NSCLC [
38]. Similarly, the combination of targeted therapies with immune checkpoint inhibitors or CAR-T cell therapies is being explored to achieve durable responses in hard-to-treat cancers [
39].
Multi-Omics Integration
Multi-omics approaches—combining genomics, transcriptomics, proteomics, and metabolomics—are providing deep insights into the molecular underpinnings of cancer. The integration of these data allows for a more comprehensive understanding of tumor biology, aiding in the identification of novel biomarkers and therapeutic targets [
40]. AI and machine learning tools are playing a crucial role in analyzing multi-omics data, improving our ability to predict patient responses to specific treatments and personalize therapies more effectively [
41].
6. Overcoming Challenges in Advanced Cancer Therapies
Tumor Heterogeneity and Drug Resistance
Tumor heterogeneity—variability within and between tumors—presents a significant challenge to cancer therapy. The clonal evolution of tumors leads to the emergence of drug-resistant subpopulations that evade treatment [
42]. Combining therapies that target different mechanisms of action can help overcome resistance by addressing both sensitive and resistant clones [
43].
Side Effects and Toxicity
Although targeted therapies and immunotherapies generally have lower toxicity than traditional chemotherapy, they are not without side effects. For example, immune checkpoint inhibitors can lead to immune-related adverse events (irAEs), ranging from mild inflammation to life-threatening autoimmune conditions [
44]. Similarly, CAR-T cell therapies are associated with cytokine release syndrome (CRS) and neurotoxicity, which require careful monitoring and management [
45]. Ongoing research aims to minimize these adverse effects while optimizing therapeutic efficacy.
7. The Role of Artificial Intelligence and Machine Learning
AI and machine learning are increasingly being integrated into cancer treatment to enhance drug discovery, optimize treatment regimens, and predict patient responses. AI algorithms are capable of analyzing large, complex datasets to uncover hidden patterns that might otherwise be missed by traditional methods [
46]. For example, machine learning models are being used to predict patient responses to immunotherapy based on tumor genomic and proteomic data, thus enabling more personalized treatment strategies [
47]. These advances in AI-driven medicine hold great promise for transforming the way cancer is treated, making therapies more precise, efficient, and accessible.
8. Future Directions and Conclusion
Ongoing Innovations
The future of cancer therapy lies in the continued integration of advanced technologies. Emerging fields such as epigenetic therapies, CRISPR-based gene editing, and tumor organoids are providing new avenues for treatment. Epigenetic therapies aim to modify gene expression without altering the underlying DNA sequence, offering a promising strategy to reverse drug resistance and enhance treatment efficacy [
48,
49]. CRISPR/Cas9-based gene editing allows for precise modifications to the genome, presenting the potential to correct genetic mutations at the root of cancers, and even engineer immune cells to target tumors more effectively [
50]. Tumor organoids, which are 3D cultures of tumor cells, are enabling better in vitro modeling of cancer and improving our understanding of tumor biology and therapeutic response [
51]. Furthermore, the development of next-generation CAR-T cells, which can target solid tumors, is another key area of research. Current CAR-T therapies have proven remarkably effective in hematologic malignancies, but expanding their use to solid tumors remains a challenge due to the immunosuppressive tumor microenvironment and antigen heterogeneity [
52].
Global Impact
While these advancements offer significant potential, their accessibility remains a challenge, particularly in low- and middle-income countries (LMICs). The high cost of antibody-mediated therapies, precision treatments, and CAR-T therapies limits their widespread use. Additionally, the infrastructure required to deliver these therapies—such as specialized centers, skilled personnel, and advanced diagnostics—is often lacking in many regions. Expanding the reach of personalized cancer therapies will require both technological innovation and global collaboration to ensure that these life-saving treatments are available to all patients. Efforts to reduce treatment costs, improve manufacturing processes, and develop mobile health solutions could play a pivotal role in democratizing access to next-generation cancer therapies [
53].
Author Contributions
Conceptualisation: JM, VBSK; Manuscript drafting: AKM; Proofread & Edit: AKM.
Acknowledgments
• ICMR, Govt. of India; • KSCSTE, Govt. of Kerala.
References
- Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-674.
- Vogelstein B, et al. Cancer genome landscapes. Science. 2013;339(6127):1546-1558. [CrossRef]
- Sawyers CL. Targeted cancer therapy. Nature. 2004;432(7015):294-297.
- Shulman H, et al. Monoclonal antibody therapy in cancer: Mechanisms and clinical applications. Oncologist. 2002;7(6):531-538.
- Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372(9):793-795. [CrossRef]
- Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443-2454. [CrossRef]
- Weiner LM, et al. Monoclonal antibodies for the treatment of cancer. Nat Med. 2006;12(5):511-518.
- van der Bij GJ, et al. Antibody-dependent cellular cytotoxicity (ADCC) in cancer therapy. Cancer Biol Ther. 2006;5(11):1315-1324.
- Lambert JM, et al. Antibody-drug conjugates for cancer therapy. Pharmacol Ther. 2014;140(1):44-55.
- Rituximab, a monoclonal antibody, is used in combination with chemotherapy to treat lymphoma and chronic lymphocytic leukemia. (Study: Tuscano, et al., J Clin Oncol. 2004).
- Tuscano, JM, et al. Rituximab in combination with chemotherapy for B-cell lymphoma and CLL. J Clin Oncol. 2004;22(9):1796-1803.
- Blinatumomab: A bispecific antibody targeting both T-cells and tumor cells. Cancer Cell. 2015;28(3):421-424.
- Lonberg, N. Bispecific antibodies for cancer immunotherapy. Nat Biotechnol. 2015;33(10):1135-1146.
- Grupp SA, et al. Chimeric antigen receptor T cells for acute lymphoblastic leukemia. N Engl J Med. 2013;368(16):1509-1518.
- Brentjens RJ, et al. Chimeric antigen receptor T-cell therapy for solid tumors. Nat Med. 2013;19(2):351-357.
- Frigault MJ, et al. Mechanisms of resistance to monoclonal antibodies. J Immunol. 2014;192(3):1501-1509.
- Lee DW, et al. Cytokine release syndrome (CRS) and neurotoxicity in CAR T-cell therapy. Cancer J. 2019;25(3):215-221.
- Neelapu SS, et al. Chimeric antigen receptor T-cell therapy: Cost considerations. Cancer. 2017;123(9):1459-1467.
- Mardis ER, et al. Next-generation DNA sequencing for cancer. Genome Research. 2009;19(11):1724-1730.
- Herbst RS, et al. EGFR inhibitors in non-small-cell lung cancer. N Engl J Med. 2004;350(21):2333-2341.
- Chapman PB, et al. BRAF inhibitors in melanoma. N Engl J Med. 2011;364(26):2507-2516.
- Druker BJ, et al. Imatinib in chronic myelogenous leukemia. N Engl J Med. 2001;344(14):1031-1037.
- Schuler M, et al. Resistance mechanisms to tyrosine kinase inhibitors in cancer therapy. Cancer Lett. 2015;367(1):58-64.
- Le DT, et al. Mismatch repair deficiency and microsatellite instability as predictors of response to PD-1 blockade in cancer. N Engl J Med. 2015;372(26):2509-2520.
- Diaz LA, et al. The molecular evolution of colorectal cancer and its implications for therapy. J Clin Oncol. 2012;30(15):1707-1713.
- Hugo W, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in melanoma. Cell. 2016;165(1):35-44. [CrossRef]
- McLemore EC, et al. Liquid biopsy for cancer: Methods and applications. J Mol Diagn. 2018;20(2):154-167.
- Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-264.
- Sharma P, et al. Immune checkpoint inhibitors: A new era in cancer treatment. Lancet Oncol. 2016;17(5):e293-e303.
- Postow MA, et al. Nivolumab and ipilimumab: Combination checkpoint blockade for cancer treatment. J Clin Oncol. 2015;33(28):3291-3299.
- Brahmer JR, et al. Nivolumab vs docetaxel in advanced non-small cell lung cancer. N Engl J Med. 2015;373(17):1627-1639.
- Kono K, et al. Immunotherapy-related adverse events in cancer patients. J Clin Oncol. 2017;35(6):401-409.
- Maude SL, et al. Chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449-459.
- Gattinoni L, et al. Immunotherapy for solid tumors. Nat Rev Immunol. 2013;13(9): 679-690.
- Biswas SK, et al. Oncolytic virus therapy in cancer treatment. Viruses. 2013;5(10):2434-2447.
- Hu K, et al. Oncolytic virotherapy in combination with CAR-T cell therapy for cancer treatment. Cancer Gene Ther. 2019;26(3):133-142.
- Schuster SJ, et al. Blinatumomab: A bispecific T-cell engager for the treatment of leukemia. Cancer. 2017;123(19):3887-3896.
- Reiss KA, et al. Combination therapy of chemotherapy and immune checkpoint inhibitors in metastatic NSCLC. Oncol. 2017;22(12):1504-1511.
- Czerwinski DK, et al. Combination targeted therapies with CAR-T and immune checkpoint inhibitors in cancer. J Clin Oncol. 2020;38(2):118-125.
- Rappoport JZ. Molecular profiling in cancer: An overview. Trends Cancer. 2017;3(7):325-334.
- Wang F, et al. AI-based analysis of multi-omics data for cancer treatment. Cancer Cell. 2020;37(6):824-830.
- Gerlinger M, et al. Intratumor heterogeneity and the evolution of cancer. Nature. 2012;486(7403):411-419.
- Sharma SV, et al. Resistance mechanisms to targeted cancer therapy. Cancer Cell. 2010;18(1):79-87.
- Weber JS, et al. Adverse effects of immune checkpoint inhibitors. Lancet Oncol. 2017;18(3):413-423.
- Lee DW, et al. Cytokine release syndrome (CRS) and neurotoxicity in CAR T-cell therapy. Cancer J. 2019;25(3):215-221.
- Rajkomar A, et al. Scalable and accurate deep learning for electronic health records. NPJ Digit Med. 2018;1(1):18. [CrossRef]
- Park YJ, et al. Predicting response to immunotherapy using machine learning. Sci Transl Med. 2019;11(488):eaav6706.
- Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683-692.
- Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346(6213):1258096. [CrossRef]
- Clevers H. Organoids: development and applications. Cell. 2016;165(7):1586-1597.
- June CH, O'Connor RS, Kawalekar OU, et al. CAR T cell therapy: the first year in review. J Clin Oncol. 2018;36(17):1697-1706.
- Alwan A, King L. The economic burden of cancer: implications for global health policy. Lancet. 2017;389(10080):1011-1020.
- Ginsburg O, Bray F, Coleman MP, et al. The global burden of cancer: disparities in incidence, mortality, and survival. CA Cancer J Clin. 2020;70(1):13-32.
Table 1.
Mechanisms of Action of Monoclonal Antibodies (mAbs).
Table 1.
Mechanisms of Action of Monoclonal Antibodies (mAbs).
| Mechanism |
Example |
Target |
Reference |
| Direct inhibition of tumor cell growth |
Trastuzumab |
HER2 (human epidermal growth factor receptor 2) |
[7] |
| Antibody-dependent cellular cytotoxicity (ADCC) |
Rituximab |
CD20 (B-cell marker) |
[8] |
| Complement-dependent cytotoxicity (CDC) |
Cetuximab |
EGFR (epidermal growth factor receptor) |
[7] |
| Antibody-drug conjugates (ADCs) |
Trastuzumab emtansine |
HER2 |
[9] |
Table 2.
Key Advances in Antibody-Mediated Cancer Therapies.
Table 2.
Key Advances in Antibody-Mediated Cancer Therapies.
| Therapy |
Target |
Mechanism of Action |
Clinical Use |
Reference |
| Monoclonal Antibodies (mAbs) |
Various (HER2, CD20, EGFR, etc.) |
Direct tumor cell targeting, ADCC, CDC, ADCs |
Breast cancer, lymphoma, etc. |
[4] |
| Bispecific Antibodies (BsAbs) |
CD19, CD3 |
T-cell and tumor cell engagement to enhance immune response |
Acute lymphoblastic leukemia, lymphoma |
[12] |
| Chimeric Antigen Receptor T-cell (CAR-T) |
CD19, BCMA |
T-cells engineered to target tumor cells |
Leukemia, lymphoma, multiple myeloma |
[14] |
Table 3.
Examples of Targeted Therapies in Precision Medicine.
Table 3.
Examples of Targeted Therapies in Precision Medicine.
| Therapy |
Target |
Cancer Type |
Clinical Impact |
Reference |
| EGFR Inhibitors |
EGFR (epidermal growth factor receptor) |
Non-small cell lung cancer (NSCLC) |
Improved survival and response rates |
[20] |
| BRAF Inhibitors |
BRAF |
Melanoma |
Enhanced progression-free survival |
[21] |
| Tyrosine Kinase Inhibitors (TKIs) |
BCR-ABL (Philadelphia chromosome) |
Chronic myelogenous leukemia (CML) |
Targeted treatment with minimal side effects |
[22] |
| ALK Inhibitors |
ALK (anaplastic lymphoma kinase) |
NSCLC (ALK-positive) |
Significant clinical benefit in advanced stages of NSCLC |
[23] |
Table 4.
Emerging Biomarkers and Their Role in Predicting Response to Cancer Therapies.
Table 4.
Emerging Biomarkers and Their Role in Predicting Response to Cancer Therapies.
| Biomarker |
Cancer Type |
Therapeutic Implication |
Reference |
| Microsatellite Instability (MSI) |
Colorectal cancer, endometrial cancer |
Predicts response to immune checkpoint inhibitors |
[24] |
| Tumor Mutational Burden (TMB) |
Various cancers |
Indicates likelihood of response to immunotherapy |
[26] |
| Circulating Tumor DNA (ctDNA) |
Various cancers |
Used for monitoring disease progression and minimal residual disease |
[25] |
Table 5.
Clinical Successes and Challenges in Immunotherapy.
Table 5.
Clinical Successes and Challenges in Immunotherapy.
| Immunotherapy |
Cancer Type |
Clinical Success |
Challenges |
Reference |
| Immune Checkpoint Inhibitors (PD-1/PD-L1, CTLA-4 inhibitors) |
Melanoma, lung cancer, bladder cancer |
Improved survival in advanced stages |
Immune-related adverse events (irAEs), resistance |
[28,29] |
| Chimeric Antigen Receptor T-cell (CAR-T) |
Leukemia, lymphoma |
High remission rates in hematologic cancers |
Cytokine release syndrome (CRS), neurotoxicity |
[33] |
| Oncolytic Viruses |
Melanoma, glioblastoma |
Selective tumor lysis and immune activation |
Limited solid tumor efficacy, tumor heterogeneity |
[35] |
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).