Cancer immunotherapy
Cancer immunotherapy is a form of biological therapy that utilizes the body's immune system to recognize, target, and eliminate cancer cells, often by enhancing natural immune responses or introducing engineered components to overcome cancer's evasion tactics.[1] Unlike traditional treatments such as chemotherapy or radiation, which directly attack rapidly dividing cells, immunotherapy leverages immune mechanisms like T cells and antibodies to provide potentially long-lasting protection against cancer recurrence.[1] The field traces its origins to the late 19th century, when surgeon William B. Coley observed tumor regressions in patients treated with bacterial toxins, leading to his development of "Coley's toxins" as an early immunotherapeutic approach that achieved remissions in over 1,000 cases of sarcoma and other cancers.[2] Key theoretical foundations emerged in the mid-20th century, including Frank Macfarlane Burnet's 1957 immunosurveillance hypothesis, which posited that the immune system constantly patrols for and eliminates nascent cancer cells, and Jacques Miller's 1967 discovery of T cells as central orchestrators of adaptive immunity.[2] Modern immunotherapy gained momentum in the 1970s with the approval of bacillus Calmette-Guérin (BCG) for bladder cancer in 1976 and the identification of the first tumor antigen in 1991, paving the way for targeted therapies.[2] Major types of cancer immunotherapy include immune checkpoint inhibitors, which block proteins like PD-1 and CTLA-4 to unleash T-cell attacks on tumors; adoptive cell therapies such as CAR T-cell therapy, where patient T cells are genetically modified to express chimeric antigen receptors targeting specific cancer markers; monoclonal antibodies that bind to cancer cell surfaces or immune modulators; treatment vaccines that stimulate immune recognition of tumor antigens; and immune system modulators like cytokines (e.g., interferons and interleukins) that amplify immune activity.[1] These approaches have been approved by the U.S. Food and Drug Administration (FDA) for over 30 cancer types, including melanoma, lung cancer, and leukemia, with notable milestones such as the 1997 approval of rituximab for non-Hodgkin lymphoma, the 2011 approval of ipilimumab as the first checkpoint inhibitor, and the 2017 approval of the first CAR T-cell therapy for pediatric acute lymphoblastic leukemia.[1][2] Recent advances underscore immunotherapy's expanding role, with the 2018 Nobel Prize in Physiology or Medicine awarded to James Allison and Tasuku Honjo for their discoveries in checkpoint inhibition that revolutionized treatment for previously intractable cancers.[2] In 2024, the FDA approved lifileucel (Amtagvi), the first tumor-infiltrating lymphocyte (TIL) therapy for advanced melanoma, and afamitresgene autoleucel, the first T-cell receptor therapy for synovial sarcoma, demonstrating durable responses in solid tumors.[3] In 2025, the FDA approved the combination of nivolumab and ipilimumab as first-line therapy for adults with unresectable or metastatic dMMR or MSI-H colorectal cancer.[4] Clinical trials have also shown neoadjuvant immunotherapy achieving complete responses without additional surgery in rectal cancer patients with mismatch repair deficiency and improving survival in kidney cancer as adjuvant therapy.[5] Despite challenges like immune-related side effects and resistance mechanisms, ongoing research into combination therapies, biomarkers for response prediction, and microbiome influences continues to broaden immunotherapy's efficacy across diverse cancers.[1]Fundamentals
Role of the Immune System in Cancer
The immune surveillance hypothesis, first articulated by Frank Macfarlane Burnet in 1957, proposes that the immune system acts as a vigilant sentinel, continuously recognizing and destroying nascent cancer cells through the detection of abnormal antigens arising from genetic mutations or cellular transformations. This concept evolved from early observations of immune tolerance and was later formalized in the framework of cancer immunoediting, which encompasses three phases: elimination of transformed cells, equilibrium where the immune system restrains tumor growth, and escape where tumors evade detection. Central to this process is the dual arm of immunity—in innate and adaptive—working synergistically to maintain tissue homeostasis and suppress oncogenesis.[6] Innate immune components provide the first line of defense against cancer cells. Natural killer (NK) cells, a subset of innate lymphoid cells, patrol peripheral tissues and rapidly lyse tumor cells that downregulate major histocompatibility complex class I (MHC-I) molecules or express stress-induced ligands such as MICA/MICB, which bind activating receptors like NKG2D on NK cells. Macrophages, another key innate player, engulf and destroy abnormal cells via phagocytosis, often triggered by pattern recognition receptors that detect damage-associated molecular patterns (DAMPs) released by dying tumor cells; they also bridge to adaptive immunity by processing and presenting antigens. These mechanisms ensure early eradication of precancerous lesions without prior sensitization.[7][8] Adaptive immunity refines this response through antigen-specific recognition, primarily orchestrated by T and B lymphocytes. Cytotoxic CD8+ T cells identify tumor antigens presented on the cell surface via MHC-I molecules by professional antigen-presenting cells (APCs) like dendritic cells, leading to targeted killing through perforin and granzyme release. Helper CD4+ T cells, activated by MHC-II presentation of exogenous antigens, coordinate responses by secreting cytokines that amplify NK and macrophage activity or support B cell differentiation into antibody-producing plasma cells, which opsonize tumor cells for destruction. Tumor antigens driving these responses are classified into tumor-associated antigens (TAAs), which are overexpressed in cancers but also present in normal tissues (e.g., HER2 in breast cancer), and tumor-specific antigens (TSAs), including neoantigens generated by somatic mutations unique to the tumor, offering highly immunogenic targets with minimal off-tumor effects.[9][10][11] Despite these protective mechanisms, tumors frequently evade immune detection through multifaceted strategies that disrupt recognition and effector functions. A common tactic is the downregulation or loss of MHC-I expression, rendering tumor cells invisible to CD8+ T cells while potentially increasing susceptibility to NK cells, though tumors often counteract this by secreting soluble MHC-I or upregulating inhibitory ligands. Additionally, tumors induce an immunosuppressive microenvironment by expressing programmed death-ligand 1 (PD-L1), which engages PD-1 on T cells to inhibit their activation and promote exhaustion. Recruitment of regulatory T cells (Tregs), characterized by FoxP3 expression, suppresses effector T cell proliferation via IL-10 and TGF-β secretion, while myeloid-derived suppressor cells (MDSCs) deplete arginine and produce reactive oxygen species to impair T cell signaling and survival. These evasion tactics collectively foster tumor progression by subverting both innate and adaptive arms of immunity.[12][13][14]Principles of Immunotherapy
Cancer immunotherapy represents a therapeutic approach that modulates the host immune system to recognize and eliminate malignant cells, distinguishing it from conventional treatments such as chemotherapy, which directly target and kill rapidly proliferating tumor cells through cytotoxic mechanisms.[1][15] By leveraging the specificity and memory capabilities of the adaptive immune system, immunotherapy seeks to restore or enhance natural antitumor immunity that has been suppressed by tumor evasion strategies, such as altered antigen presentation or inhibitory signaling.[15] The core principles of cancer immunotherapy revolve around four interrelated strategies: improving antigen presentation to prime immune recognition, amplifying effector cell functions to mount a robust attack, alleviating tumor-induced immunosuppression to sustain immune activity, and promoting the development of long-term immunological memory to prevent recurrence. Enhancing antigen presentation involves strategies that increase the visibility of tumor-associated antigens to antigen-presenting cells like dendritic cells, thereby initiating T-cell activation. Boosting effector cells, such as cytotoxic T lymphocytes and natural killer cells, focuses on expanding their numbers and potency to directly lyse tumor targets. Relieving immunosuppression targets mechanisms that dampen immune responses, allowing unchecked effector function. Finally, inducing memory responses ensures that surviving immune cells, particularly memory T cells, provide durable protection against residual or metastatic disease.[16][15] Key concepts underpinning these principles include the abscopal effect and epitope spreading, which illustrate the potential for systemic and broadening immune responses. The abscopal effect describes the regression of non-irradiated tumors following localized therapy, driven by radiation-induced release of tumor antigens that trigger a widespread adaptive immune response via activated T cells.[17] Epitope spreading refers to the diversification of the antitumor immune response, where initial targeting of specific tumor antigens leads to recognition of additional, previously ignored epitopes, thereby broadening the immune attack and reducing the risk of antigen escape.[18] Ultimately, the goals of cancer immunotherapy are to achieve sustained, durable clinical responses, particularly in immunogenic or "hot" tumors characterized by high immune cell infiltration and antigen burden, which respond more readily than "cold" tumors with sparse immune presence and low immunogenicity. In hot tumors, therapies can amplify existing immune pressure for complete remissions, whereas in cold tumors, the challenge lies in converting an immunosuppressive microenvironment to one conducive to effective immunity, often requiring combination approaches to ignite initial responses.[19][20]Historical Development
Early Observations and Concepts
The earliest observations linking infections to cancer regression date back to the late 19th century, when physicians noted spontaneous remissions in tumors following acute febrile illnesses, suggesting an immune-mediated response to microbial stimuli.[21] These anecdotal reports gained traction through the work of surgeon William B. Coley, who in 1891 observed a patient with inoperable sarcoma whose tumor regressed after developing erysipelas, a streptococcal infection.[22] Inspired by this, Coley developed "Coley's toxins," a mixture of heat-killed Streptococcus pyogenes and Serratia marcescens bacteria, which he administered to over 1,000 cancer patients starting in 1893; in some cases, particularly sarcomas, tumors regressed, with long-term remissions reported in up to 20% of treated individuals, though mechanisms were then attributed to fever and inflammation rather than specific immunity.[23][24] In the early 1900s, Paul Ehrlich advanced theoretical foundations for targeted cancer therapies with his "magic bullet" concept, proposing that specific antibodies could selectively bind and destroy pathogens or abnormal cells, much like a directed projectile.[25] Ehrlich extended this to cancer, hypothesizing in his side-chain theory that the immune system recognizes altered self-antigens on tumor cells and that vaccines could enhance this recognition, laying groundwork for immunotherapeutic ideas despite initial focus on infectious diseases.[26] By the mid-20th century, the immune surveillance theory formalized the notion that the immune system continuously monitors and eliminates nascent cancer cells. In 1957, Frank Macfarlane Burnet proposed this concept, arguing that lymphocytes detect and destroy mutant cells expressing neoantigens, preventing tumor formation as a natural evolutionary adaptation.[27] Lewis Thomas extended the theory in 1959, emphasizing the role of cellular immunity in recognizing tumor-specific antigens and integrating it with broader immunological principles, influencing subsequent research on immune-cancer interactions.[28] Concurrent discoveries highlighted soluble immune factors with anticancer potential. In 1957, Alick Isaacs and Jean Lindenmann identified interferon, a protein secreted by virus-infected cells that inhibits viral replication and later demonstrated antitumor effects in preclinical models by activating immune responses.[29] Building on bacterial immunostimulation concepts, early 1970s experiments by Alvaro Morales explored intravesical Bacillus Calmette-Guérin (BCG), an attenuated tuberculosis vaccine; in a 1976 trial of nine bladder cancer patients, BCG reduced tumor recurrence rates dramatically compared to historical controls, marking the first clinical evidence of bacterial immunotherapy's efficacy.[30]Key Milestones and Regulatory Approvals
The development of hybridoma technology in 1975 by Georges Köhler and César Milstein revolutionized antibody production by enabling the creation of monoclonal antibodies through the fusion of antibody-producing B cells with myeloma cells, allowing continuous culture of hybrid cells secreting antibodies of predefined specificity. This breakthrough earned them the Nobel Prize in Physiology or Medicine in 1984, laying the foundation for antibody-based cancer immunotherapies. Building briefly on early 19th-century observations such as William Coley's use of bacterial toxins to stimulate immune responses against tumors, these advancements shifted immunotherapy toward targeted, reproducible interventions.[31] The first major regulatory milestone came in 1997 with the FDA approval of rituximab (Rituxan), the inaugural monoclonal antibody for cancer treatment, indicated for relapsed or refractory low-grade or follicular CD20-positive non-Hodgkin lymphoma, marking the entry of targeted antibody therapies into clinical practice. Subsequent approvals expanded the field, including pembrolizumab (Keytruda) in 2014 as the first PD-1 inhibitor for unresectable or metastatic melanoma, demonstrating durable responses in checkpoint blockade immunotherapy. That same year, blinatumomab (Blincyto) received accelerated approval as the first bispecific T-cell engager antibody for relapsed or refractory Philadelphia chromosome-negative precursor B-cell acute lymphoblastic leukemia, redirecting T cells to tumor cells. Adoptive cell therapies achieved pivotal approvals in 2017, with the FDA granting approval to tisagenlecleucel (Kymriah), the first CAR-T cell therapy, for pediatric and young adult patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Shortly after, axicabtagene ciloleucel (Yescarta) was approved for relapsed or refractory large B-cell lymphoma, establishing CAR-T as a transformative approach for hematologic malignancies. Earlier, in 2011, ipilimumab (Yervoy) became the first checkpoint inhibitor approved by the FDA for unresectable or metastatic melanoma, based on phase III trial data showing improved overall survival. Recent years have seen further diversification, with elranatamab (Elrexfio) approved in 2023 for relapsed or refractory multiple myeloma after at least four prior lines of therapy, representing a bispecific antibody targeting BCMA and CD3. In 2024, lifileucel (Amtagvi) received accelerated FDA approval as the first tumor-infiltrating lymphocyte (TIL) therapy for adult patients with unresectable or metastatic melanoma previously treated with a PD-1 inhibitor and targeted therapy if BRAF V600 mutation-positive. In May 2025, the FDA approved nivolumab plus ipilimumab as first-line therapy for unresectable or metastatic microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) colorectal cancer, based on phase 3 CheckMate 8HW trial data demonstrating improved progression-free survival and overall survival.[4] Post-2020, mRNA-based cancer vaccines have advanced through key clinical trials, including Moderna's mRNA-4157 entering phase III in 2022 for high-risk melanoma in combination with pembrolizumab, showing promising immune activation and tumor response rates in earlier phases. Progress in TIGIT inhibitors has been highlighted by phase 2 data from 2024, such as the EDGE-Gastric trial where domvanalimab plus zimberelimab and chemotherapy demonstrated a median progression-free survival exceeding one year in first-line unresectable or metastatic gastric, gastroesophageal junction, or esophageal adenocarcinoma, though other candidates like Roche's tiragolumab faced setbacks in liver cancer studies.[32]| Year | Therapy | Class | Indication | Source |
|---|---|---|---|---|
| 1997 | Rituximab (Rituxan) | Monoclonal antibody | Relapsed/refractory CD20+ non-Hodgkin lymphoma | FDA Approval History |
| 2011 | Ipilimumab (Yervoy) | CTLA-4 inhibitor | Unresectable/metastatic melanoma | FDA Approval |
| 2014 | Pembrolizumab (Keytruda) | PD-1 inhibitor | Unresectable/metastatic melanoma | FDA Approval |
| 2014 | Blinatumomab (Blincyto) | Bispecific antibody | Relapsed/refractory Ph- B-cell ALL | FDA Approval |
| 2017 | Tisagenlecleucel (Kymriah) | CAR-T cell therapy | Relapsed/refractory B-cell ALL (pediatric/young adult) | FDA Approval |
| 2017 | Axicabtagene ciloleucel (Yescarta) | CAR-T cell therapy | Relapsed/refractory large B-cell lymphoma | FDA Approval |
| 2023 | Elranatamab (Elrexfio) | Bispecific antibody | Relapsed/refractory multiple myeloma (≥4 prior lines) | FDA Approval |
| 2024 | Lifileucel (Amtagvi) | TIL therapy | Unresectable/metastatic melanoma (post-PD-1/targeted therapy) | FDA Approval |
| 2025 | Nivolumab + ipilimumab | Checkpoint inhibitor combination | First-line MSI-H/dMMR unresectable/metastatic colorectal cancer | FDA Approval |
Cellular Immunotherapies
Dendritic Cell-Based Therapies
Dendritic cells (DCs) serve as professional antigen-presenting cells that capture, process, and present tumor antigens to T cells, thereby initiating and amplifying adaptive immune responses against cancer.[33] In DC-based therapies, autologous DCs are harvested from the patient's peripheral blood via leukapheresis, cultured ex vivo to expand and mature them, loaded with tumor-specific antigens such as peptides, tumor lysates, or mRNA, and then reinfused to prime antitumor T-cell responses.[33] This approach leverages the DCs' ability to migrate to lymph nodes and activate naive T cells, offering a personalized immunotherapy strategy that avoids off-the-shelf limitations while minimizing graft-versus-host risks.[34] A landmark example is sipuleucel-T (Provenge), the first FDA-approved DC-based vaccine for metastatic castration-resistant prostate cancer in 2010. In its manufacturing process, peripheral blood mononuclear cells are isolated from leukapheresis products, and monocytes are differentiated into DCs using granulocyte-macrophage colony-stimulating factor (GM-CSF); these DCs are then activated by co-culture with the fusion protein prostatic acid phosphatase (PAP)-GM-CSF, matured with cytokines including interleukin-1β, tumor necrosis factor-α, and interleukin-6, and infused back into the patient in three biweekly doses.[35] The pivotal phase 3 IMPACT trial, involving 512 patients, demonstrated a 22% reduction in the risk of death (hazard ratio 0.78; 95% CI, 0.62 to 0.98), with median overall survival of 25.8 months versus 21.7 months in the placebo group, establishing sipuleucel-T's efficacy in extending survival without significant toxicity.[35] Beyond sipuleucel-T, other DC-based approaches include pulsing autologous DCs with tumor-specific peptides, such as those derived from melanoma-associated antigens like gp100 or NY-ESO-1, followed by maturation and intradermal or intravenous administration.[36] In advanced melanoma, phase 2 trials have shown that peptide-pulsed DC vaccines can induce antigen-specific T-cell responses and objective responses in up to 10-20% of patients, with some achieving durable progression-free survival when combined with checkpoint inhibitors.[37] For glioblastoma, DCs electroporated with tumor mRNA, such as Wilms' tumor 1 (WT1) mRNA, have been tested in phase 1/2 trials, demonstrating safety, immunogenicity through WT1-specific T-cell expansion, and median overall survival of 15-18 months in recurrent cases post-standard therapy.[38] Despite these advances, DC-based therapies face challenges including suboptimal DC maturation, which can impair antigen presentation and T-cell priming, and limited migration to draining lymph nodes, with studies reporting only 1-5% of injected mature DCs reaching lymphoid tissues in melanoma patients.[39] These issues contribute to variable clinical efficacy, prompting ongoing research into adjuvants like toll-like receptor agonists to enhance maturation and chemokine modifications to improve lymph node homing.[40]Adoptive Cell Transfer Therapies
Adoptive cell transfer therapies involve the extraction, ex vivo modification, and reinfusion of a patient's own immune cells to enhance their tumor-targeting capabilities, primarily focusing on T cells and natural killer (NK) cells engineered for direct cytotoxicity against cancer. These approaches leverage the patient's autologous cells to minimize rejection risks while amplifying anti-tumor responses, often following lymphodepleting chemotherapy to create a favorable microenvironment for engraftment. Unlike indirect activation methods, such as dendritic cell therapies that prime systemic immunity, adoptive transfers deliver pre-activated cells directly into circulation for immediate tumor engagement.[41] Tumor-infiltrating lymphocyte (TIL) therapy extracts T cells from a patient's resected tumor, expands them in vitro using interleukin-2 to enrich for tumor-reactive clones, and reinfuses them to target heterogeneous tumor antigens. This method harnesses naturally occurring TILs that have already infiltrated the tumor microenvironment, recognizing multiple neoantigens for broader efficacy against antigen escape. In February 2024, the FDA granted accelerated approval to lifileucel (Amtagvi), the first TIL therapy, for adults with unresectable or metastatic melanoma previously treated with other therapies, based on an objective response rate of 31.4% in a phase 2 trial.[42][43][44] Chimeric antigen receptor (CAR) T-cell therapy engineers patient T cells to express synthetic CARs, which combine an antigen-binding domain—typically a single-chain variable fragment from an antibody—with intracellular signaling domains from CD3ζ and costimulatory molecules like CD28 or 4-1BB to activate cytotoxicity upon target engagement. Initially developed for B-cell malignancies, CD19-targeted CAR-T cells have shown durable remissions; for instance, tisagenlecleucel (Kymriah) received FDA approval in August 2017 for relapsed or refractory pediatric and young adult B-cell acute lymphoblastic leukemia, achieving a 82% complete remission rate in pivotal trials. Similarly, axicabtagene ciloleucel (Yescarta) was approved in October 2017 for large B-cell lymphoma, with a 72% objective response rate and 52% complete responses.[45][46][47] Expansion of CAR-T therapy to solid tumors targets surface antigens like GD2 in neuroblastoma and H3K27M-mutated gliomas, or HER2 in breast and gastric cancers, though challenges include tumor heterogeneity and immunosuppressive microenvironments. Phase 1 trials of GD2-CAR-T cells in H3K27M-altered diffuse midline gliomas reported clinical improvements and tumor reductions in 9 of 11 patients, with some durable responses exceeding two years. HER2-targeted CAR-T therapies have demonstrated feasibility in solid tumors, with ongoing trials showing partial responses in gastric and colorectal cancers without severe on-target toxicities when using lower-affinity binders.[48][49][50] T-cell receptor (TCR) T-cell therapy genetically engineers peripheral blood T cells to express tumor-specific TCRs that recognize intracellular antigens presented on MHC class I molecules, enabling targeting of neoantigens unique to cancer cells. This approach is particularly suited for solid tumors, where neoantigens arise from mutations; clinical trials have tested TCR-T cells against public neoantigens like KRAS G12D in colorectal and pancreatic cancers, achieving objective responses in up to 33% of patients with minimal off-tumor toxicity. Ongoing phase 1/2 studies focus on personalized neoantigen-specific TCRs identified via tumor sequencing, with preliminary data showing T-cell persistence and tumor infiltration in melanoma and synovial sarcoma.[51][52][53] NK cell therapies, including CAR-NK and cytokine-induced killer (CIK) cells, offer advantages over T-cell approaches by avoiding graft-versus-host disease (GVHD) due to the absence of TCR-mediated alloreactivity, allowing potential allogeneic "off-the-shelf" use. CAR-NK cells express CARs targeting tumor antigens like CD19 or HER2, combined with IL-15 for enhanced persistence; early trials in lymphoma and solid tumors report complete remissions without cytokine release syndrome in some patients. CIK cells, generated by culturing peripheral blood mononuclear cells with interferon-γ and IL-2, exhibit a mixed T/NK phenotype with non-MHC-restricted killing, showing safety and modest efficacy in hepatocellular carcinoma when combined with chemotherapy.[54][55][56]Antibody-Based Therapies
Monoclonal Antibodies
Monoclonal antibodies (mAbs) represent a cornerstone of antibody-based cancer immunotherapy, consisting of unmodified or minimally modified immunoglobulin molecules designed to bind specific tumor-associated antigens or immune regulators on cancer cells. These naked mAbs, lacking chemical conjugates or bispecific engineering, harness the immune system to target and eliminate malignant cells while sparing healthy tissues. Approved examples include rituximab, a chimeric anti-CD20 mAb approved by the U.S. Food and Drug Administration (FDA) on November 26, 1997, for the treatment of relapsed or refractory CD20-positive, B-cell low-grade or follicular non-Hodgkin's lymphoma.[57] Similarly, trastuzumab, a humanized anti-HER2 mAb, received FDA approval on September 25, 1998, for patients with metastatic breast cancer whose tumors overexpress the HER2 protein and who have undergone prior chemotherapy or, in combination with paclitaxel, for those without prior metastatic chemotherapy.[58] More recent approvals include dinutuximab, a chimeric anti-GD2 mAb approved on March 10, 2015, for pediatric patients with high-risk neuroblastoma following initial multimodal therapy.[59] The therapeutic efficacy of these naked mAbs primarily arises from three key mechanisms: antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and direct blockade of tumor-promoting signaling pathways. In ADCC, the Fc region of the mAb binds to Fcγ receptors on effector cells such as natural killer cells, recruiting them to lyse antibody-coated tumor cells. CDC involves the activation of the complement system upon antigen binding, forming a membrane attack complex that perforates the tumor cell membrane. Direct signaling blockade occurs when mAbs bind to receptors like HER2, inhibiting ligand-induced proliferation signals in cancer cells. For instance, rituximab targets the CD20 antigen on B-cell lymphomas, primarily inducing cell death via ADCC and CDC. Trastuzumab blocks HER2 dimerization and downstream signaling in breast cancer cells, complemented by ADCC. Dinutuximab binds the GD2 glycolipid on neuroblastoma cells, triggering lysis through ADCC and CDC.[59] In investigational settings, magrolimab, a humanized anti-CD47 mAb, disrupts the CD47-SIRPα interaction to remove the "don't eat me" signal on cancer cells, thereby promoting macrophage-mediated phagocytosis; it received FDA breakthrough therapy designation in 2020 for untreated intermediate- or high-risk myelodysplastic syndrome; however, development was discontinued in 2024 following a clinical hold due to safety concerns.[60] To mitigate immunogenicity associated with early murine-derived mAbs, which elicited human anti-mouse antibody responses limiting repeated dosing, humanization techniques evolved progressively. Chimeric antibodies, such as rituximab developed in the 1990s, fused murine variable regions to human constant regions, reducing immunogenicity while retaining antigen specificity.[61] Humanized mAbs like trastuzumab, approved shortly thereafter, further minimized murine content by grafting only the complementarity-determining regions (CDRs) onto human frameworks, enhancing tolerability and efficacy in clinical use.[61] The advent of phage display technology in 1990 enabled the selection of fully human antibodies from large synthetic or immune libraries, bypassing animal immunization and yielding candidates with near-native human sequences; this approach underpins later oncology mAbs, though its first major cancer application came with approvals like panitumumab in 2006 targeting EGFR in colorectal cancer.[61]Bispecific Antibodies and Conjugates
Bispecific antibodies (BsAbs) represent an advanced class of engineered immunotherapeutics designed to simultaneously bind two distinct antigens or epitopes, thereby enhancing tumor targeting and immune activation in cancer treatment. Unlike traditional monoclonal antibodies, BsAbs facilitate dual functionality, such as redirecting T cells to tumor cells or blocking multiple pathways, which has led to improved efficacy in hematologic malignancies and emerging applications in solid tumors. These molecules are typically constructed using formats like single-chain variable fragments or full-length IgG scaffolds to achieve stable heterodimerization.[62] A prominent subclass of BsAbs includes T-cell engagers, which link tumor-associated antigens on cancer cells to CD3 on T cells, promoting cytotoxic synapse formation and tumor cell lysis. Blinatumomab, a CD19/CD3 bispecific T-cell engager, was granted accelerated FDA approval in December 2014 for relapsed or refractory B-cell precursor acute lymphoblastic leukemia (ALL) in adults and children, demonstrating a complete remission rate of 44% in pivotal trials.[63] More recently, elranatamab, a BCMA/CD3 bispecific antibody, received accelerated FDA approval in August 2023 for relapsed or refractory multiple myeloma after at least four prior lines of therapy, with an objective response rate of 61% observed in the MagnetisMM-1 study.[64] These approvals highlight the clinical impact of T-cell engagers in redirecting endogenous T cells without prior ex vivo manipulation.[65] Antibody-drug conjugates (ADCs) extend the immunotherapy paradigm by conjugating monoclonal antibodies to cytotoxic payloads via chemical linkers, enabling targeted delivery of chemotherapeutics to tumor cells while minimizing systemic toxicity. Trastuzumab deruxtecan, an HER2-targeted ADC with a topoisomerase I inhibitor payload, was approved by the FDA in December 2019 under accelerated approval for unresectable or metastatic HER2-positive breast cancer following prior anti-HER2 therapies, based on a confirmed objective response rate of 37.1% in the DESTINY-Breast01 trial.[66] In January 2025, the FDA expanded approval of fam-trastuzumab deruxtecan for unresectable or metastatic hormone receptor-positive, HER2-low or ultralow breast cancer previously treated with endocrine therapy and chemotherapy.[67] Similarly, sacituzumab govitecan, a Trop-2-directed ADC linked to SN-38 (an irinotecan metabolite), received accelerated FDA approval in April 2020 for metastatic triple-negative breast cancer after two prior therapies, with median progression-free survival of 5.6 months versus 1.7 months for single-agent chemotherapy in the ASCENT trial; full approval followed in April 2021.[68] These ADCs leverage linker stability and bystander killing effects to enhance potency against heterogeneous tumors.[69] Beyond ADCs, other conjugates include radioimmunoconjugates, where antibodies are labeled with radionuclides to deliver targeted radiation. Ibritumomab tiuxetan, an anti-CD20 antibody conjugated to yttrium-90 or indium-111, was approved by the FDA in 2002 for relapsed or refractory low-grade or follicular non-Hodgkin lymphoma, offering improved response rates when combined with rituximab compared to rituximab alone.[70] Bispecific T-cell engagers targeting non-CD3 moieties, such as NKG2D or CD16, are also under investigation to broaden immune effector engagement.[71] Engineering strategies for BsAbs often employ the knob-into-hole (KiH) technology, introduced in 1996, which introduces specific mutations in the Fc region's CH3 domains—one chain with a bulky "knob" residue (e.g., T366W) and the other with a complementary "hole" (e.g., T366S/L368A/Y407V)—to favor heterodimer assembly over homodimerization, achieving over 95% correct pairing in production.[72] This approach has facilitated scalable manufacturing of clinical-grade BsAbs. In solid tumors, BsAbs are demonstrating clinical advantages in ongoing trials as of 2025, including enhanced tumor infiltration and reduced escape mechanisms; for instance, phase II studies of EGFR/CD3 engagers report objective response rates up to 40% in advanced colorectal cancer, with multiple trials advancing to registrational intent.[73][74]Immune Checkpoint Blockade
CTLA-4 and PD-1/PD-L1 Inhibitors
Immune checkpoint inhibitors targeting CTLA-4 represent a foundational advance in cancer immunotherapy by augmenting early T-cell activation. Ipilimumab, a fully human monoclonal antibody against CTLA-4, was approved by the U.S. Food and Drug Administration (FDA) on March 25, 2011, as the first therapy for unresectable or metastatic melanoma, marking the initial demonstration of prolonged survival with checkpoint blockade.[75] CTLA-4, expressed on activated T cells, competes with the co-stimulatory receptor CD28 for binding to B7 ligands on antigen-presenting cells, thereby dampening T-cell priming in lymph nodes; ipilimumab blocks this interaction to promote robust T-cell proliferation and anti-tumor responses.[76] This mechanism primarily acts in secondary lymphoid tissues during the priming phase of the immune response, distinguishing it from other checkpoints. PD-1 inhibitors disrupt a key inhibitory pathway in the tumor microenvironment, reinvigorating exhausted T cells. Nivolumab, an IgG4 monoclonal antibody, received FDA approval on December 22, 2014, for the treatment of unresectable or metastatic melanoma after progression on ipilimumab and, if BRAF V600 mutation-positive, a BRAF inhibitor.[77] Similarly, pembrolizumab, a humanized IgG4 antibody, was approved on September 4, 2014, for ipilimumab-refractory advanced melanoma.[77] Both agents bind directly to PD-1 on the surface of T cells, preventing engagement with its ligands PD-L1 and PD-L2, which tumors exploit to suppress cytotoxic activity and foster immune evasion.[78] PD-L1 inhibitors complement PD-1 blockade by targeting the ligand often overexpressed on tumor cells and associated immune cells. Atezolizumab, a humanized IgG1 monoclonal antibody, became the first PD-L1 inhibitor approved by the FDA on May 18, 2016, for locally advanced or metastatic urothelial carcinoma progressing after platinum-containing chemotherapy.[77] Durvalumab, another human IgG1κ antibody, followed with FDA approval on May 1, 2017, for platinum-refractory advanced or metastatic urothelial carcinoma.[77] These inhibitors bind to PD-L1, blocking its interaction with PD-1 on T cells and CD80 on antigen-presenting cells, thereby relieving T-cell suppression within the tumor niche.[79] These agents have yielded durable clinical responses across malignancies, with ipilimumab demonstrating a doubling of 10-year overall survival rates in metastatic melanoma compared to historical controls.[80] In non-small cell lung cancer, PD-1 and PD-L1 inhibitors like nivolumab and atezolizumab have produced objective response rates of 20-30% and extended progression-free survival in advanced cases, often with responses lasting beyond two years.[81] Combinations, such as ipilimumab plus nivolumab, further improve outcomes, achieving 5-year survival rates exceeding 50% in melanoma.[82] Predictive biomarkers include microsatellite instability-high (MSI-H) status, which correlates with higher response rates due to increased neoantigen load, and elevated tumor mutational burden (TMB), where high-TMB tumors show hazard ratios for progression-free survival as low as 0.47 with PD-1 blockade.[83][84]Novel Checkpoint Targets
Immune checkpoints beyond CTLA-4 and PD-1/PD-L1, such as LAG-3, TIGIT, TIM-3, and VISTA, have emerged as promising targets in cancer immunotherapy to overcome resistance mechanisms, particularly in immunologically "cold" tumors characterized by low T-cell infiltration and poor response to PD-1 blockade.[85] These molecules are often co-expressed on exhausted T cells in the tumor microenvironment, where they sustain immunosuppression even after PD-1 inhibition, providing a rationale for dual or multi-checkpoint blockade strategies to reinvigorate antitumor immunity.[86] By targeting these pathways, therapies aim to enhance T-cell activation and infiltration in resistant tumors, with ongoing trials evaluating combinations to broaden immunotherapy efficacy across solid malignancies.[87] LAG-3 (lymphocyte-activation gene 3) is an inhibitory receptor expressed on activated and exhausted CD8+ T cells and regulatory T cells, where it interacts with major histocompatibility complex class II to dampen T-cell proliferation and cytokine production, contributing to immune evasion in tumors.[88] The first LAG-3 inhibitor, relatlimab, a human IgG4 monoclonal antibody, was approved by the FDA in March 2022 as part of the fixed-dose combination Opdualag (relatlimab plus nivolumab) for adult and pediatric patients (aged 12 and older) with unresectable or metastatic melanoma, based on the phase 3 RELATIVITY-047 trial demonstrating a median progression-free survival of 10.1 months versus 4.6 months with nivolumab alone (hazard ratio 0.75).[89][90][91] As of 2025, LAG-3 combinations continue in trials for other indications, though the phase 3 RELATIVITY-098 adjuvant study in resected melanoma missed its primary endpoint for disease-free survival when added to nivolumab.[92] TIGIT (T-cell immunoreceptor with Ig and ITIM domains) functions as an inhibitory receptor on T cells and natural killer cells, competing with the costimulatory receptor DNAM-1 for ligands like CD155 and CD112 to suppress cytotoxic responses and promote regulatory T-cell activity in the tumor microenvironment.[93] Tiragolumab, an anti-TIGIT monoclonal antibody, has been investigated in combination with atezolizumab (anti-PD-L1), showing early promise in the phase 2 CITYSCAPE trial for non-small cell lung cancer (NSCLC) with an objective response rate of 37% versus 21% for atezolizumab alone.[93] However, phase 3 results as of 2025 have been disappointing: the SKYSCRAPER-01 trial reported no significant improvement in progression-free survival (median 7.0 months versus 5.6 months) or overall survival with the addition of tiragolumab in PD-L1-high untreated NSCLC, while SKYSCRAPER-02 similarly failed to show benefit in extensive-stage small cell lung cancer when combined with atezolizumab and chemotherapy (median PFS 5.4 months versus 5.6 months).[94][95][96] TIM-3 (T-cell immunoglobulin and mucin-domain containing-3) is another exhaustion marker on T cells and natural killer cells, binding ligands like galectin-9 to induce apoptosis and inhibit IFN-γ production, often upregulated alongside PD-1 in resistant tumors to perpetuate T-cell dysfunction.[97] As of 2025, TIM-3 inhibitors remain in early clinical development, with the phase 1 trial of INCAGN02390 (a fully human anti-TIM-3 antibody) demonstrating tolerability and preliminary antitumor activity as monotherapy or in combination with PD-1 inhibitors across advanced solid tumors, including reduced exhausted CD8+ T cells in preclinical models.[98][99] Ongoing trials, such as combinations with PD-1 blockade in melanoma and lung cancer, aim to address resistance by enhancing T-cell infiltration, with phase 1b/2 data supporting triplet regimens including LAG-3 for improved event-free survival.[100][101] VISTA (V-domain Ig suppressor of T-cell activation) acts as an inhibitory ligand on myeloid cells and T cells, suppressing T-cell proliferation via VSIG-3 interaction and promoting an immunosuppressive microenvironment, particularly in acidic tumor conditions that favor its expression.[102] In 2025, VISTA-targeted therapies are primarily in preclinical and early-phase trials, with monoclonal antibodies showing enhanced antitumor effects in combination with PD-1/PD-L1 inhibitors in models of triple-negative breast cancer and pancreatic cancer by blunting radiotherapy-induced myeloid suppression.[103][104] Bispecific antibodies targeting VISTA and PD-L1 have demonstrated superior tumor inhibition in preclinical studies of endometrial and breast cancers compared to monotherapies, supporting ongoing phase 1 evaluations for broader application in immunotherapy-resistant settings.[105][106]Cytokine-Based Therapies
Interferons
Interferons are a family of cytokines that play a pivotal role in cancer immunotherapy by modulating immune responses and directly inhibiting tumor growth. Type I interferons, including IFN-α and IFN-β, bind to a common receptor to activate the JAK-STAT signaling pathway, which induces the expression of genes promoting antiviral states, apoptosis, and immune cell activation. This pathway specifically enhances natural killer (NK) cell cytotoxicity and T-cell proliferation, contributing to antitumor effects in various malignancies.[107][108] Type II interferon, IFN-γ, signals through a distinct receptor but also engages JAK-STAT to upregulate major histocompatibility complex (MHC) class I molecules on tumor cells, improving antigen presentation to cytotoxic T cells, while simultaneously inhibiting angiogenesis by downregulating vascular endothelial growth factor (VEGF) expression.[109][110] IFN-α has been a cornerstone of cytokine-based cancer therapy since its recombinant form was first approved by the U.S. Food and Drug Administration (FDA) in 1986 for hairy cell leukemia, where it induces tumor cell differentiation and immune-mediated clearance, achieving response rates of up to 80% in early studies. In 1995, high-dose IFN-α-2b was approved as adjuvant therapy for high-risk resected melanoma, demonstrating improvements in relapse-free survival by 9-12 months in pivotal trials through enhanced NK and T-cell activity via JAK-STAT signaling. Pegylated formulations, such as peginterferon alfa-2b (Sylatron), were later approved in 2011 for adjuvant melanoma treatment, offering sustained release with a longer half-life (approximately 40 hours [range 22-60 hours] versus ~5 hours for standard IFN-α), which reduces dosing frequency from daily to weekly and improves patient compliance while maintaining efficacy in preventing recurrence.[111][112][113][114] These pegylated versions have also been combined with chemotherapy agents like dacarbazine in metastatic melanoma, showing synergistic effects by sensitizing tumor cells to apoptosis and boosting immune surveillance, with response rates around 20-30% in phase II trials.[115][116] IFN-β and IFN-γ have been explored primarily in investigational settings, particularly for brain tumors like glioblastoma. IFN-β inhibits glioma angiogenesis by suppressing VEGF production and has shown modest activity in phase II trials for recurrent glioblastoma, with some patients achieving stable disease for over 6 months when administered intratumorally or systemically. IFN-γ upregulates MHC class I on glioblastoma cells, potentially enhancing T-cell recognition, and preclinical studies indicate it curbs tumor vascularization; however, clinical trials in high-grade gliomas have yielded mixed results, with limited survival benefits when added to standard temozolomide chemotherapy, prompting ongoing research into optimized delivery methods.[117][118][119] Prior to the advent of immune checkpoint inhibitors in the 2010s, interferons represented one of the earliest approved immunotherapies, with IFN-α serving as a standard for hematologic and solid tumors from the 1980s onward, though its use declined due to toxicity profiles. Common side effects include flu-like symptoms such as fever, chills, fatigue, and myalgias, affecting up to 70% of patients, alongside hematologic toxicities like leukopenia and elevated liver enzymes, which are often dose-dependent and manageable with supportive care or dose reductions.[120][121][122]Interleukins
Interleukins represent a class of cytokines pivotal in cancer immunotherapy, particularly for their role in stimulating immune effector cells against tumors. Among them, interleukin-2 (IL-2), marketed as aldesleukin, has been a cornerstone therapy due to its ability to drive the expansion and activation of T lymphocytes and natural killer (NK) cells, thereby enhancing antitumor immune responses. High-dose IL-2 administration promotes robust proliferation of CD8+ cytotoxic T cells and NK cells by binding to the high-affinity IL-2 receptor (IL-2Rαβγ), leading to downstream signaling via JAK/STAT pathways that amplify effector functions and cytokine production.[123] The U.S. Food and Drug Administration (FDA) approved high-dose aldesleukin in 1992 for metastatic renal cell carcinoma and in 1998 for metastatic melanoma, based on clinical trials demonstrating durable complete responses in approximately 5-10% of patients, though with significant toxicity including vascular leak syndrome and hypotension requiring intensive monitoring.[124] Dosing typically involves intravenous bolus infusions of 600,000-720,000 IU/kg every 8 hours for up to 14 doses per cycle, with toxicity management strategies such as premedication with antihistamines and fluid support to mitigate capillary permeability issues.[125] Low-dose IL-2 regimens have emerged as a strategy to preferentially expand regulatory T cells (Tregs) expressing high levels of CD25 (IL-2Rα), aiming to modulate the immunosuppressive tumor microenvironment while minimizing systemic toxicity. At doses around 1-3 million IU/m² subcutaneously, low-dose IL-2 sustains Treg populations to prevent autoimmunity but, in cancer settings, has shown potential to enhance effector T-cell function when combined with immune checkpoint inhibitors like PD-1 blockers.[126] Ongoing trials from 2020-2025, such as those combining low-dose IL-2 with nivolumab in melanoma and renal cell carcinoma, report improved progression-free survival rates (e.g., up to 40% at 12 months in select cohorts) by balancing immune activation and suppression, though challenges remain in optimizing dosing to avoid over-stimulation of Tregs.[127] Toxicity at these levels is generally mild, limited to flu-like symptoms, contrasting sharply with high-dose regimens.[128] Beyond IL-2, other interleukins like IL-15 and IL-12 are under investigation for their complementary roles in effector cell expansion and immune polarization. IL-15 agonists, such as NKTR-255—a pegylated IL-15 receptor agonist—selectively stimulate CD8+ T cells and NK cells without activating Tregs, showing promise in phase 1/2 trials through 2025 when combined with CAR-T therapies for B-cell malignancies, where complete response rates reached 70-80% at 6 months versus 50% with CAR-T alone.[129] IL-12 drives a Th1 immune shift by inducing interferon-γ (IFN-γ) production from NK and T cells, promoting cytotoxic responses and angiogenesis inhibition, with intratumoral delivery strategies in recent trials demonstrating tumor regression in solid tumors like melanoma without severe systemic effects.[130] Clinical studies, including phase 1 evaluations of recombinant IL-12, highlight its efficacy in shifting the cytokine milieu toward Th1 dominance, though dose-limiting hepatotoxicity has prompted localized administration approaches.[131] Advances as of 2025 focus on engineered IL-2 variants to enhance specificity and curb toxicity, such as tumor-targeted immunocytokines fusing IL-2 to antibodies against tumor-associated antigens like PD-1 or CD8β. These variants, including nemvaleukin alfa (an IL-2 mutein lacking α-receptor binding), achieve selective delivery to the tumor site, reducing vascular leak while boosting intratumoral T-cell infiltration and yielding objective response rates of 20-30% in phase 2 trials for advanced solid tumors when paired with checkpoint inhibitors.[132] Combinatorial strategies, such as co-administration with JAK inhibitors like upadacitinib, further abrogate systemic exposure, enabling safer high-potency dosing and improved tolerability in ongoing studies.[133]Oncolytic Virus Therapy
Mechanisms of Action
Oncolytic viruses (OVs) are genetically modified or naturally occurring viruses that selectively infect and replicate within tumor cells, leading to their lysis and the release of tumor antigens and danger signals that stimulate an anti-tumor immune response. This dual mechanism distinguishes OVs from traditional chemotherapies by combining direct cytotoxicity with immunogenic modulation of the tumor microenvironment. The selectivity of OVs for cancer cells arises from inherent tumor vulnerabilities, such as defective antiviral signaling pathways, and targeted genetic engineering to attenuate virulence in normal cells while preserving replication in malignant ones.[134] A key example of engineered selectivity is seen in talimogene laherparepvec (T-VEC), a modified herpes simplex virus type 1 (HSV-1) where both copies of the ICP34.5 gene are deleted. The ICP34.5 protein normally counters host antiviral responses by dephosphorylating eIF2α, but its absence restricts viral replication in healthy cells with intact interferon signaling; in contrast, tumor cells often exhibit dysregulated RAS pathways or impaired protein kinase R (PKR) activation, allowing selective viral propagation and subsequent cell lysis. Similarly, the oncolytic adenovirus H101 features a deletion in the E1B-55K gene, enabling replication in p53-deficient cancer cells—common in nasopharyngeal carcinoma—while sparing normal p53-proficient cells; H101 received approval in China in 2005 for this indication. Reoviruses, such as reolysin, demonstrate natural oncolysis without genetic modification, exploiting activated oncogenic pathways like RAS to bypass the need for junction oncogene adhesion molecule A (JAM-A) in normal cells, thus preferentially infecting and lysing transformed cells.[135][136][137] Beyond direct tumor cell destruction, OVs induce immunogenic cell death (ICD), releasing damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1) from lysed cells, which acts as a danger signal to recruit and activate innate immune cells. HMGB1 binds to receptors like TLR4 on dendritic cells (DCs), promoting their maturation and enhancing cross-presentation of tumor-associated antigens to CD8+ T cells, thereby priming adaptive anti-tumor immunity; this process briefly leverages DC antigen presentation mechanisms to amplify systemic responses. The resulting inflammation transforms immunologically "cold" tumors into "hot" ones, fostering T-cell infiltration and long-term immune memory.[138][139] While the primary effects of OV therapy are local—confined to injected tumor sites—the immune activation often yields systemic benefits, including abscopal responses where uninjected distant metastases regress due to primed T-cell trafficking. Preclinical and early clinical observations with viruses like T-VEC and adenoviral OVs have demonstrated these abscopal effects, underscoring the potential for broad anti-tumor activity beyond direct lysis.[140][134]Clinical Examples and Applications
Talimogene laherparepvec (T-VEC), a modified herpes simplex virus type 1, was approved by the U.S. Food and Drug Administration in 2015 for the local treatment of unresectable cutaneous, subcutaneous, and nodal melanoma in patients with no evidence of visceral metastases.[141] Administered via intralesional injection, T-VEC selectively replicates in tumor cells, leading to direct oncolysis and induction of systemic antitumor immunity. In the phase III OPTiM trial (NCT00769704), involving 436 patients with advanced melanoma, T-VEC demonstrated a durable response rate (DRR) of 16.3% compared to 2.1% with subcutaneous granulocyte-macrophage colony-stimulating factor (GM-CSF), with median overall survival of 23.3 months versus 18.9 months, respectively.[142] The therapy's safety profile was favorable, with common adverse events including fatigue, chills, and injection-site reactions, and no treatment-related deaths reported.[143] H101 (Oncorine), an E1B-55K gene-deleted oncolytic adenovirus, became the world's first approved oncolytic virus therapy when it received approval from China's State Food and Drug Administration in 2005 for the treatment of advanced head and neck cancer, particularly nasopharyngeal carcinoma, in combination with cisplatin and 5-fluorouracil chemotherapy.[144] Delivered via intratumoral injection, H101 targets p53-deficient tumor cells common in these cancers, enhancing chemotherapy efficacy through viral replication and immune activation. Phase III clinical trials in China showed that the combination improved objective response rates to 78.8% compared to 39.6% with chemotherapy alone, with median survival extended to 12.1 months versus 9.9 months, and a tolerable safety profile dominated by flu-like symptoms and mild injection-site pain.[145] H101 has been used in numerous patients in China since approval, underscoring its established role in this indication.[145] Pexa-Vec (pexastimogene devacirepvec), a thymidine kinase-deleted vaccinia virus engineered to express GM-CSF, has been investigated for advanced hepatocellular carcinoma (HCC) and other solid tumors but faced setbacks in late-stage development. The phase III PHOCUS trial (NCT02562755), evaluating intravenous Pexa-Vec followed by sorafenib in 600 patients with advanced HCC, was terminated early in 2019 after an interim futility analysis showed no improvement in overall survival (median 12.7 months [95% CI: 9.89-14.95] for combination versus 14.0 months [95% CI: 11.01-17.22] for sorafenib alone), with inferior outcomes in key subgroups.[146] Despite this failure, data from a phase I/II study in refractory metastatic colorectal cancer combining intravenous Pexa-Vec with durvalumab (anti-PD-L1) and/or tremelimumab (anti-CTLA-4) showed objective response rates of up to 6.25% and disease control rates of 12.5-16.7%, with no unexpected toxicities beyond grade 1-2 flu-like symptoms.[147] Ongoing trials continue to explore these synergies, particularly in immunologically "cold" tumors like HCC.[148] Oncolytic virus therapies, including T-VEC and H101, have primarily found applications in accessible solid tumors such as melanoma, head and neck cancers, and HCC, where direct injection facilitates tumor targeting and immune priming.[149] Combinations with PD-1 inhibitors have shown additive effects by enhancing T-cell infiltration and systemic responses; for instance, T-VEC plus pembrolizumab in neoadjuvant melanoma yielded a pathologic complete response rate of 44% in a phase II trial.[150] Similarly, integration with CAR-T cell therapy addresses solid tumor barriers like immunosuppression, with preclinical and early clinical data indicating that oncolytic viruses precondition the tumor microenvironment to boost CAR-T persistence and efficacy, achieving up to 80% tumor regression in mouse models of glioma and ovarian cancer.[151] Emerging examples include cretostimogene grenadenorepvec, an oncolytic adenovirus showing promising phase 3 results (BOND-003 trial) for high-risk BCG-unresponsive non-muscle-invasive bladder cancer, with a complete response rate of 75.2% at any time and durability in 51% of responders at 12 months as of 2025, pending U.S. FDA approval following a planned Biologics License Application submission in Q4 2025.[152] These approaches are advancing toward broader use in refractory solid malignancies, emphasizing multimodal strategies to overcome monotherapy limitations.[153]Cancer Vaccines
Preventive and Therapeutic Vaccines
Preventive vaccines in cancer immunotherapy target oncogenic viruses to inhibit cancer development before it occurs. The human papillomavirus (HPV) vaccine Gardasil, approved by the FDA in 2006, prevents cervical, vulvar, vaginal, and anal cancers caused by HPV types 16 and 18 by inducing neutralizing antibodies that block viral entry into host cells. Clinical trials demonstrated over 90% efficacy in preventing HPV-related precancerous lesions in women aged 9-26. Similarly, the hepatitis B virus (HBV) vaccine, recommended by the CDC and WHO since the 1980s, prevents chronic HBV infection, which is a major risk factor for hepatocellular carcinoma, reducing liver cancer incidence by up to 70% in vaccinated populations through the generation of protective anti-HBs antibodies. These vaccines exemplify how immunization against viral carcinogens can significantly lower cancer rates at the population level. Therapeutic vaccines, in contrast, aim to elicit immune responses against established tumors by presenting tumor-associated antigens to stimulate antigen-specific T cells and antibodies. They induce both humoral immunity, via B-cell production of tumor-targeting antibodies, and cellular immunity, through CD8+ cytotoxic T lymphocytes that recognize and lyse cancer cells. Adjuvants such as granulocyte-macrophage colony-stimulating factor (GM-CSF) enhance these responses by recruiting and activating dendritic cells at the vaccination site, promoting antigen processing and presentation. These mechanisms differ from dendritic cell-based therapies, which involve ex vivo manipulation of patient-derived cells but share the goal of boosting adaptive immunity. Examples of therapeutic vaccines include whole-cell approaches like GVAX for prostate cancer, which uses irradiated allogeneic prostate cancer cell lines (LNCaP and PC-3) engineered to secrete GM-CSF, thereby presenting multiple tumor antigens to induce broad T-cell responses; phase II trials showed immune activation but mixed survival benefits. Viral vector vaccines, such as PROSTVAC, employ recombinant poxviruses (vaccinia prime followed by fowlpox boosts) encoding prostate-specific antigen (PSA) and costimulatory molecules (TRICOM), leading to PSA-specific T-cell proliferation; a phase II trial in metastatic castration-resistant prostate cancer reported an 8.5-month overall survival extension versus placebo, though a subsequent phase III trial failed to meet its primary endpoint of statistical significance for overall survival.[154] Polysaccharide vaccines like Polysaccharide-K (PSK), derived from Trametes versicolor and approved in Japan in 1977 as an adjuvant for gastric cancer, activate innate immunity via TLR2 agonism, enhancing NK cell and T-cell activity to prolong disease-free survival when combined with chemotherapy.Personalized Neoantigen Vaccines
Personalized neoantigen vaccines are designed to elicit immune responses against tumor-specific mutations unique to an individual's cancer, offering a tailored approach to immunotherapy. Neoantigens arise from somatic mutations in tumor DNA, and their identification begins with next-generation sequencing (NGS) of tumor and matched normal tissue to detect these alterations, followed by human leukocyte antigen (HLA) typing to determine how neoantigens are presented on the cell surface.[155] Bioinformatics algorithms then predict peptide binding to HLA molecules and assess potential immunogenicity by evaluating factors such as binding affinity, stability, and similarity to self-antigens to prioritize candidates likely to trigger T-cell responses.[156] Advanced machine learning models integrate proteogenomic data to rank neoantigens, improving accuracy in selecting those with high therapeutic potential.[156] Vaccine platforms for delivering these neoantigens vary, with mRNA-based approaches enabling rapid, individualized manufacturing by encoding selected neoantigen sequences into lipid nanoparticles for direct translation in dendritic cells. For instance, BNT111, an mRNA vaccine targeting four melanoma-associated neoantigens, has been evaluated in phase II trials for advanced melanoma, often combined with PD-1 inhibitors to enhance efficacy.[157] Similarly, Moderna's mRNA-4157 (also known as V940), which encodes up to 34 patient-specific neoantigens, initiated phase III trials starting in 2023 for adjuvant therapy in high-risk melanoma, with a phase III trial for non-small cell lung cancer beginning in 2024; phase II data indicated a relapse-free survival rate of approximately 75% at 18 months, and 3-year follow-up as of 2024 showed a sustained 49% reduction in the risk of recurrence or death compared to pembrolizumab alone.[158] Peptide-based platforms, such as NeoVax, use synthetic long peptides mixed with adjuvants like poly-ICLC to mimic natural antigen processing, targeting up to 20 neoantigens per patient in melanoma settings.[159] Clinical trials combining personalized neoantigen vaccines with checkpoint inhibitors have shown promising immune activation and antitumor activity, particularly in melanoma. In the phase II BNT111-01 trial, BNT111 plus cemiplimab achieved an objective response rate of 18.1% in PD-(L)1-refractory melanoma patients, with vaccine-specific T-cell responses correlating to clinical benefit.[160] For mRNA-4157 combined with pembrolizumab in resected high-risk melanoma, phase II data indicated a relapse-free survival rate of approximately 75% at 18 months, highlighting the synergy of neoantigen targeting with immune checkpoint blockade.[161] NeoVax trials in stage III/IV melanoma reported durable T-cell responses and a median progression-free survival of over two years in small cohorts, underscoring the approach's potential for long-term control.[162] These vaccines hold particular promise for tumors with low tumor mutational burden (TMB), where shared antigens are limited, as neoantigens can still be derived from non-coding mutations, splice variants, or frameshifts to drive specific immunity without off-target effects.[163] By 2025, advances in artificial intelligence have enhanced epitope prediction, with deep learning models achieving higher accuracy in forecasting HLA presentation and immunogenicity, reducing false positives and accelerating vaccine design for broader applicability.[164] Such AI integrations, including transformer-based architectures, enable more precise selection of neoantigens even in immunologically "cold" tumors.[165]Biomarkers and Patient Selection
Genetic and Molecular Biomarkers
Genetic and molecular biomarkers play a crucial role in predicting responses to cancer immunotherapy by identifying tumor-intrinsic alterations that influence immune recognition and checkpoint inhibitor efficacy. Microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) tumors exhibit enhanced responsiveness to PD-1/PD-L1 inhibitors due to increased neoantigen load and immune activation. In 2017, the U.S. Food and Drug Administration (FDA) granted accelerated approval for pembrolizumab in unresectable or metastatic MSI-H or dMMR solid tumors, marking the first tissue-agnostic approval based on this biomarker, with subsequent companion diagnostics like the MMR IHC 4-in-1 pharmDx and FoundationOne CDx enabling its clinical use. Similarly, tumor mutational burden (TMB), defined as the number of nonsynonymous mutations per megabase of genome, serves as a proxy for neoantigen immunogenicity; tumors with TMB greater than 10 mutations per megabase (mut/Mb) demonstrate higher objective response rates to immune checkpoint inhibitors across various cancers, including non-small cell lung cancer and melanoma, though thresholds may vary by tumor type.[166][167][168][169] Certain genetic alterations confer resistance to immunotherapy, highlighting the need for targeted genomic profiling. Loss-of-function mutations in JAK1 or JAK2 disrupt interferon-gamma signaling, impairing antigen presentation and leading to primary or acquired resistance to PD-1 blockade in melanomas and other solid tumors. In melanoma, BRAF V600 mutations, present in approximately 50% of cases, influence immunotherapy outcomes; while these tumors respond to checkpoint inhibitors, BRAF status guides sequential or combinatorial strategies with targeted BRAF/MEK inhibitors to enhance immune infiltration and response durability. Recent research as of 2025 has identified loss-of-function mutations in PPP2R1A, a gene encoding a protein phosphatase subunit, as associated with significantly prolonged overall and progression-free survival in patients receiving immunotherapy, particularly in ovarian clear cell carcinoma and high-risk endometrial cancer, suggesting potential as a novel positive predictive biomarker. These findings underscore the prognostic value of specific gene mutations in stratifying patients for immunotherapy.[170][171][172][173][174] Next-generation sequencing (NGS) panels are the primary method for detecting these biomarkers, enabling comprehensive tumor profiling. The MSK-IMPACT assay, a hybridization capture-based NGS panel targeting over 400 cancer-associated genes, accurately quantifies TMB and identifies MSI status by comparing tumor and matched normal DNA, facilitating precision oncology decisions for immunotherapy eligibility. As of 2025, liquid biopsy techniques using circulating tumor DNA (ctDNA) have advanced TMB assessment, offering non-invasive monitoring; studies demonstrate that blood-based TMB correlates with tissue TMB and predicts progression-free survival in immunotherapy-treated patients, though standardization of cutoffs remains a challenge for broader adoption.[175][176][177][178]Tumor Microenvironment and Immune Biomarkers
The tumor microenvironment (TME) plays a pivotal role in modulating immune responses to cancer, with biomarkers assessing its composition providing critical insights for predicting immunotherapy efficacy. These markers evaluate the balance between immune activation and suppression within the tumor ecosystem, distinguishing immunologically "hot" tumors—characterized by robust T-cell infiltration and responsiveness to checkpoint inhibitors—from "cold" tumors with sparse immune engagement. Key TME biomarkers include surface protein expression on tumor cells, densities of infiltrating immune cells, suppressive elements, and even distal influences like the gut microbiome, which collectively inform patient stratification for therapies such as PD-1/PD-L1 inhibitors. Additionally, systemic inflammatory markers such as the neutrophil-to-lymphocyte ratio (NLR) derived from complete blood count, serum lactate dehydrogenase (LDH), and albumin levels have been recommended by the Society for Immunotherapy of Cancer (SITC) consensus in 2025 as essential for inclusion in clinical trials to predict response and toxicity across tumor types.[179][180] PD-L1 expression, assessed via immunohistochemistry (IHC), remains a cornerstone biomarker for selecting patients for anti-PD-1/PD-L1 therapies. The tumor proportion score (TPS), which quantifies the percentage of viable tumor cells exhibiting partial or complete PD-L1 membrane staining, guides eligibility for pembrolizumab in non-small cell lung cancer (NSCLC); for instance, a TPS of ≥50% identifies patients suitable for first-line monotherapy, based on FDA-approved companion diagnostics like the Dako PD-L1 IHC 22C3 pharmDx assay. This threshold correlates with improved progression-free survival in metastatic NSCLC cohorts treated with pembrolizumab plus chemotherapy, where high PD-L1 expression enhances antitumor T-cell reactivation. Automated TPS analysis has further standardized scoring, reducing inter-observer variability and confirming its prognostic value across NSCLC subtypes.[181][182][183] Immune cell infiltration patterns within the TME offer additional predictive power, particularly through metrics of cytotoxic T-cell density and organized structures. High CD8+ T-cell density in tumor cores and invasive margins pre-treatment predicts overall survival in patients receiving immune checkpoint inhibitors (ICIs), as visualized via whole-body PET imaging or multiplexed assays, reflecting an active antitumor response. Tertiary lymphoid structures (TLS)—ectopic lymphoid aggregates containing B cells, T cells, and dendritic cells—further stratify prognosis; their presence in the TME associates with favorable outcomes and enhanced ICI responses across solid tumors, including melanoma and lung cancer, by fostering local T- and B-cell priming against neoantigens. In colorectal cancer, TLS maturity (e.g., germinal center formation) correlates with reduced recurrence risk post-immunotherapy. Emerging as of October 2025, thymic health—assessed via AI-derived algorithms on CT imaging of the thymus—has been linked to improved progression-free and overall survival with immunotherapy across diverse cancers like melanoma, renal cell carcinoma, and lung cancer, highlighting the role of peripheral immune organ function in T-cell repertoire diversity and response prediction.[184][185][186][187] Suppressive elements in the TME, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), counteract immune activation and serve as inverse biomarkers of response. FOXP3+ Tregs, identified by IHC or flow cytometry, indicate an immunosuppressive milieu; however, in advanced melanoma, higher baseline intratumoral FOXP3+ Treg levels combined with TGF-β expression predict favorable anti-PD-1 responses, possibly due to heightened reliance on PD-1 blockade for Treg modulation. Elevated Ki67+ proliferating Tregs pre-treatment, conversely, signal poor event-free survival in soft tissue sarcomas under ICIs, as low levels allow better T-effector dominance post-therapy. For MDSCs, arginase-1 (ARG1) expression—measured via qPCR or IHC—marks their immunosuppressive activity by depleting L-arginine essential for T-cell function; high ARG1+ MDSC infiltration in peripheral blood or tumors correlates with reduced ICI efficacy in NSCLC and correlates with advanced disease stages.[188][189][190] The gut microbiome influences systemic immunity and TME dynamics, emerging as a non-invasive biomarker for ICI outcomes. Enrichment of Bifidobacterium species, such as B. longum or B. bifidum, in responders' fecal metagenomes associates with augmented dendritic cell function and CD8+ T-cell activation; clinical trials in melanoma patients demonstrate that oral Bifidobacterium supplementation enhances anti-PD-1 efficacy by promoting IFN-γ production and reducing Treg activity. In non-responders, dysbiosis with low Bifidobacterium diversity predicts progression, underscoring microbiome profiling via 16S rRNA sequencing as a tool for response prediction.[191][192] By 2025, advances in multiplex IHC and spatial transcriptomics have refined TME biomarker assessment, enabling precise classification of hot versus cold tumors. Multiplex IHC panels, staining up to 40 markers simultaneously, map heterogeneous immune landscapes in situ, revealing that hot tumors with clustered CD8+ T cells near tumor nests respond better to ICIs than cold tumors lacking such infiltration. Spatial transcriptomics, integrating NanoString GeoMx or Visium platforms, quantifies gene expression gradients across TME compartments, identifying immunosuppressive signatures (e.g., ARG1 upregulation in stromal regions) that predict resistance; in breast and lung cancers, these tools have validated TLS proximity to tumor edges as a superior prognostic metric over bulk RNA-seq. Such technologies facilitate personalized strategies to "heat up" cold tumors via combination therapies.[193][179][194]Approved Therapies and Clinical Use
List of FDA-Approved Agents
Cancer immunotherapy has seen numerous FDA approvals across various agent classes, beginning with monoclonal antibodies in the late 1990s and expanding to advanced cellular therapies by 2025. These agents target specific immune pathways or cancer antigens to enhance the body's antitumor response, with approvals granted for hematologic and solid tumors based on clinical trial data demonstrating efficacy and safety. The following catalogs key approved agents by class, highlighting initial approval dates and primary indications; many have received subsequent label expansions for additional cancers or settings.[195][1]Checkpoint Inhibitors
These agents block inhibitory signals on T cells, such as CTLA-4 or PD-1/PD-L1, to unleash immune attacks on tumors. Ipilimumab (Yervoy), a CTLA-4 monoclonal antibody, received FDA approval on March 25, 2011, for the treatment of unresectable or metastatic melanoma in adults and children aged 12 years and older. Pembrolizumab (Keytruda), a PD-1 inhibitor, was first approved on September 4, 2014, for unresectable or metastatic melanoma, with over 30 subsequent expansions by 2025 for cancers including non-small cell lung cancer, head and neck squamous cell carcinoma, and microsatellite instability-high solid tumors.[195] Atezolizumab (Tecentriq), a PD-L1 inhibitor, gained approval on May 18, 2016, for locally advanced or metastatic urothelial carcinoma after platinum-containing chemotherapy. Durvalumab (Imfinzi), a PD-L1 inhibitor, was approved on March 28, 2025, with gemcitabine and cisplatin as neoadjuvant treatment followed by single-agent durvalumab as adjuvant therapy for muscle-invasive bladder cancer.[196] Nivolumab (Opdivo) in combination with ipilimumab (Yervoy) was approved on April 8, 2025, for unresectable or metastatic microsatellite instability-high or mismatch repair deficient colorectal cancer.[197] Retifanlimab-dlwr (Zynyz), a PD-1 inhibitor, was approved on May 15, 2025, in combination with carboplatin and paclitaxel as first-line treatment for locally recurrent or metastatic anal squamous cell carcinoma.[198] Penpulimab-kcqx was approved on April 23, 2025, in combination with cisplatin or carboplatin and gemcitabine, and as monotherapy, for recurrent or metastatic non-keratinizing nasopharyngeal carcinoma.[199] Cemiplimab-rwlc (Libtayo), a PD-1 inhibitor, received approval on October 8, 2025, as adjuvant treatment for high-risk cutaneous squamous cell carcinoma following surgery and radiation.[200]CAR-T Cell Therapies
Chimeric antigen receptor (CAR) T-cell therapies involve engineering patient T cells to target cancer-specific antigens, primarily for hematologic malignancies. Tisagenlecleucel (Kymriah) was approved on August 30, 2017, for pediatric and young adult patients with relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Axicabtagene ciloleucel (Yescarta) received approval on October 18, 2017, for adults with relapsed or refractory large B-cell lymphoma after two or more lines of systemic therapy. Idecabtagene vicleucel (Abecma) was approved on March 26, 2021, for adult patients with relapsed or refractory multiple myeloma after four or more prior lines of therapy.Monoclonal Antibodies and Bispecific Engagers
Early immunotherapies include antibodies that bind tumor antigens or bridge immune effector cells to cancer cells. Rituximab (Rituxan), an anti-CD20 monoclonal antibody, was approved on November 26, 1997, for relapsed or refractory low-grade or follicular CD20-positive B-cell non-Hodgkin lymphoma. Trastuzumab (Herceptin), targeting HER2, received approval on December 3, 1998, as adjuvant therapy for HER2-overexpressing node-positive breast cancer. Blinatumomab (Blincyto), a bispecific T-cell engager targeting CD19 and CD3, was approved on December 3, 2014, for relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Linvoseltamab-gcpt (Lynozyfic), a bispecific BCMA-directed CD3 T-cell engager, was approved on July 2, 2025, for adults with relapsed or refractory multiple myeloma after at least four prior lines of therapy including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 monoclonal antibody.[201]Oncolytic Virus Therapies
Talimogene laherparepvec (T-VEC, Imlygic), a genetically modified herpes simplex virus, was approved on October 27, 2015, for the local treatment of melanoma lesions in patients with unresectable cutaneous, subcutaneous, or nodal lesions.Cancer Vaccines and TIL Therapies
Sipuleucel-T (Provenge), an autologous cellular vaccine, was approved on April 29, 2010, for asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. Lifileucel (Amtagvi), the first tumor-infiltrating lymphocyte (TIL) therapy, received accelerated approval on February 16, 2024, for adults with unresectable or metastatic melanoma previously treated with a PD-1 blocking antibody and targeted therapy if BRAF V600 mutation-positive.[41]Antibody-Drug Conjugates (Recent Updates)
Datopotamab deruxtecan (Datroway), a Trop-2-directed antibody-drug conjugate, was approved on January 17, 2025, for unresectable or metastatic hormone receptor-positive, HER2-negative breast cancer after endocrine therapy and prior chemotherapy, and on June 23, 2025, for previously treated locally advanced or metastatic EGFR-mutated non-small cell lung cancer.[202][203]| Class | Agent | Initial Approval Date | Primary Indication |
|---|---|---|---|
| Checkpoint Inhibitor | Ipilimumab | March 25, 2011 | Unresectable/metastatic melanoma |
| Checkpoint Inhibitor | Pembrolizumab | September 4, 2014 | Unresectable/metastatic melanoma (expansions ongoing) |
| Checkpoint Inhibitor | Atezolizumab | May 18, 2016 | Locally advanced/metastatic urothelial carcinoma |
| Checkpoint Inhibitor | Durvalumab | March 28, 2025 | Neoadjuvant/adjuvant muscle-invasive bladder cancer |
| Checkpoint Inhibitor | Nivolumab + Ipilimumab | April 8, 2025 | MSI-H/dMMR metastatic colorectal cancer |
| Checkpoint Inhibitor | Retifanlimab-dlwr | May 15, 2025 | Locally recurrent/metastatic anal squamous cell carcinoma (first-line with chemo) |
| Checkpoint Inhibitor | Penpulimab-kcqx | April 23, 2025 | Recurrent/metastatic non-keratinizing nasopharyngeal carcinoma |
| Checkpoint Inhibitor | Cemiplimab-rwlc | October 8, 2025 | Adjuvant high-risk cutaneous squamous cell carcinoma |
| CAR-T | Tisagenlecleucel | August 30, 2017 | Relapsed/refractory B-cell ALL (pediatric/young adult) |
| CAR-T | Axicabtagene ciloleucel | October 18, 2017 | Relapsed/refractory large B-cell lymphoma |
| CAR-T | Idecabtagene vicleucel | March 26, 2021 | Relapsed/refractory multiple myeloma |
| Monoclonal Antibody | Rituximab | November 26, 1997 | Relapsed/refractory non-Hodgkin lymphoma |
| Monoclonal Antibody | Trastuzumab | December 3, 1998 | HER2+ breast cancer (adjuvant) |
| Bispecific Engager | Blinatumomab | December 3, 2014 | Relapsed/refractory B-cell ALL |
| Bispecific Engager | Linvoseltamab-gcpt | July 2, 2025 | Relapsed/refractory multiple myeloma (after ≥4 lines) |
| Oncolytic Virus | Talimogene laherparepvec (T-VEC) | October 27, 2015 | Unresectable melanoma lesions |
| Vaccine | Sipuleucel-T | April 29, 2010 | Metastatic castration-resistant prostate cancer |
| TIL Therapy | Lifileucel | February 16, 2024 | Unresectable/metastatic melanoma (post-PD-1) |
| ADC | Datopotamab deruxtecan | January 17, 2025 (breast); June 23, 2025 (lung) | HR+/HER2- breast cancer; EGFR-mutated NSCLC |