Fact-checked by Grok 2 weeks ago

Checkpoint inhibitor

A checkpoint inhibitor is a type of drug, typically a , that blocks proteins on T cells or cancer cells, thereby preventing the suppression of the immune response and enabling T cells to recognize and destroy tumor cells. These inhibitors target key regulatory pathways in the , such as CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), which competes with the co-stimulatory receptor to inhibit early T cell activation, and the PD-1/PD-L1 axis ( and its ligand), which dampens T cell activity in peripheral tissues to avoid but is exploited by tumors for evasion. By disrupting these negative feedback loops, checkpoint inhibitors unleash antitumor immunity, leading to durable responses in a subset of patients across various malignancies. Approved checkpoint inhibitors include CTLA-4 blockers like , which was the first to receive FDA approval in 2011 for advanced , and PD-1 inhibitors such as and nivolumab, widely used for cancers including non-small cell lung cancer, , and . PD-L1 inhibitors like , , and target the on tumor cells, while newer agents such as (a LAG-3 combined with nivolumab) address additional checkpoints to overcome . These therapies are often administered intravenously and can be used alone or in combination with other s like or , significantly improving survival rates in responsive tumors but eliciting immune-related adverse events due to widespread T cell activation. Despite their transformative impact—earning the 2018 in Physiology or Medicine for discoveries in mechanisms—challenges persist, including primary or acquired resistance in many patients and toxicities affecting organs like the skin, colon, lungs, and endocrine glands. Ongoing research explores biomarkers for patient selection, such as expression levels, tumor mutational burden, and microsatellite instability, to optimize efficacy and minimize risks.

Introduction

Definition

Checkpoint inhibitors are a class of immunotherapy drugs, consisting of monoclonal antibodies, that block immune checkpoint proteins to enhance T-cell-mediated anti-tumor immunity. These agents target inhibitory receptors and ligands expressed on immune cells, particularly T lymphocytes, preventing the suppression of anti-tumor responses. Immune checkpoints represent a set of regulatory proteins, such as CTLA-4 and PD-1, that function as brakes on the immune system to maintain self-tolerance and prevent autoimmunity. However, tumors exploit these checkpoints by upregulating their expression or ligands, thereby dampening T-cell activation and enabling immune evasion. By inhibiting these proteins, checkpoint inhibitors restore the immune system's ability to recognize and attack cancer cells. In distinction from other immunotherapies, checkpoint inhibitors do not directly activate or engineer immune cells; unlike , which involves modifying patient T cells to express tumor-specific receptors, or therapies like interleukin-2 that broadly stimulate immune and activity, these inhibitors specifically remove suppressive signals to reinvigorate endogenous anti-tumor immunity. Most approved checkpoint inhibitors target cell surface proteins, though investigational agents are exploring intracellular checkpoints for broader modulation.

Importance in Cancer Immunotherapy

Checkpoint inhibitors have transformed by representing a from conventional treatments such as and radiation, which typically offer short-term tumor control but limited long-term efficacy, to strategies that unleash the patient's against tumors, resulting in durable responses even in advanced, previously intractable cancers. This evolution has enabled sustained remissions, with some patients achieving complete tumor eradication and long-term survival without ongoing therapy, fundamentally altering the therapeutic landscape for . As of November 2025, thirteen FDA-approved immune checkpoint inhibitors are integrated into treatment regimens for more than 20 cancer types, demonstrating response rates of 40-60% in biomarker-selected patients with melanoma and non-small cell lung cancer, far surpassing historical benchmarks with traditional agents. For instance, in advanced melanoma, these therapies have elevated 10-year overall survival rates to 43% with nivolumab plus ipilimumab combination, compared to pre-2011 five-year survival rates below 10% and median survival of just six to nine months under chemotherapy. The broader implications extend to personalized medicine, where biomarkers like PD-L1 expression guide patient selection to optimize outcomes and minimize ineffective treatments. This foundational work was underscored by the 2018 Nobel Prize in Physiology or Medicine awarded to James P. Allison and Tasuku Honjo for discovering cancer therapy through inhibition of negative immune regulation, highlighting the global impact on oncology.

Mechanism of Action

Role of Immune Checkpoints

Immune checkpoints are regulatory molecules that fine-tune T-cell responses to maintain and prevent pathological overactivation. In normal , these checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and (PD-1), act as brakes on T-cell activation during distinct phases of the . CTLA-4 primarily functions in the early priming phase within lymphoid organs, where it competes with the co-stimulatory receptor for binding to B7 ligands ( and ) on antigen-presenting cells, thereby attenuating T-cell proliferation, cytokine production like interleukin-2 (IL-2), and overall activation to avoid excessive inflammation. PD-1, in contrast, predominates during the effector phase in peripheral tissues, where engagement with its ligands PD-L1 or PD-L2 inhibits (TCR) signaling, reduces cytokine secretion such as interferon-gamma (IFN-γ), and promotes T-cell anergy or , safeguarding against . Together, these mechanisms ensure self-tolerance by downregulating responses to self-antigens and limiting tissue damage from prolonged immune activity, as evidenced by autoimmune disorders in CTLA-4- or PD-1-deficient models. In the context of cancer, tumors exploit these checkpoints to evade immune surveillance. Cancer cells frequently upregulate PD-L1 expression in response to inflammatory cytokines or oncogenic signaling, enabling it to bind PD-1 on tumor-infiltrating T cells and induce a state of T-cell exhaustion characterized by impaired effector functions and reduced antitumor . Similarly, CTLA-4 on T cells or regulatory T cells (Tregs) competes more avidly than for B7 ligands, depriving effector T cells of necessary and thereby dampening the initial anti-tumor in the . Central to these processes are key inhibitory pathways that intersect with TCR signaling. Both CTLA-4 and PD-1 recruit phosphatases like SHP-1 and SHP-2 upon ligation, dephosphorylating key TCR-associated molecules such as ZAP-70 and CD3ζ, which disrupts downstream signaling cascades including PI3K-AKT and MAPK pathways essential for T-cell activation. Additionally, Tregs constitutively express high levels of CTLA-4, which enhances their suppressive capacity by depleting B7 ligands from antigen-presenting cells through trans-endocytosis and promoting an immunosuppressive milieu that further inhibits effector T-cell responses.

How Checkpoint Inhibitors Function

Checkpoint inhibitors primarily function through monoclonal antibodies that specifically bind to immune checkpoint proteins on T cells, thereby blocking their interaction with ligands and preventing inhibitory signaling. For instance, anti-PD-1 antibodies bind to the PD-1 receptor, disrupting its engagement with or PD-L2 on tumor cells or antigen-presenting cells, which otherwise recruits phosphatases like SHP-2 to inhibit signaling. This blockade restores T-cell activation, promoting proliferation and the release of pro-inflammatory cytokines such as IFN-γ and IL-2, essential for amplifying anti-tumor responses. Similarly, anti-CTLA-4 antibodies bind CTLA-4, preventing its competition with the costimulatory receptor for B7 ligands on antigen-presenting cells, thus enhancing early T-cell priming. The therapeutic action occurs at distinct stages of the . CTLA-4 blockade predominantly enhances T-cell priming in secondary lymphoid organs like lymph nodes, where it reduces regulatory T-cell suppression and broadens the T-cell repertoire against tumor antigens. In contrast, PD-1/PD-L1 blockade acts primarily in the , reinvigorating exhausted effector T cells by alleviating chronic inhibitory signals, thereby boosting their cytotoxic function directly at the site of tumor growth. These complementary mechanisms allow checkpoint inhibitors to target both the initiation and execution phases of anti-tumor immunity. Upon blockade, an immune cascade is initiated that enhances + T-cell infiltration into tumors, improves recognition of tumor-specific antigens such as neoantigens, and fosters the formation of long-lived memory T cells. This leads to increased tumor cell and sustained immune surveillance, contributing to durable clinical responses observed in responsive patients. The reactivation of these processes disrupts tumor-induced immune suppression, enabling a more robust and persistent anti-tumor immune attack. Checkpoint inhibitors are typically administered intravenously, with half-lives ranging from approximately 6 to 27 days depending on the specific inhibitor, which supports dosing regimens often every 2-6 weeks to maintain therapeutic levels while minimizing frequent interventions. This pharmacokinetic profile arises from their large molecular size and target-mediated clearance, allowing sustained blockade of checkpoint signaling over extended periods.

Types

CTLA-4 Inhibitors

CTLA-4 inhibitors target the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), an receptor primarily expressed on activated T cells and regulatory T cells (Tregs). Upon T-cell activation, CTLA-4 competes with the co-stimulatory receptor for to (B7-1) and (B7-2) ligands on antigen-presenting cells, but with 2- to 4-fold higher affinity, delivering an inhibitory signal that attenuates early-stage T-cell priming, , and production in lymphoid tissues. By to CTLA-4 and blocking this , these monoclonal antibodies restore -mediated , thereby enhancing T-cell activation and promoting anti-tumor immune responses. A distinctive feature of CTLA-4 inhibition is its impact on Tregs, which constitutively express high levels of CTLA-4; blockade triggers Treg depletion through (ADCC), reducing immunosuppressive activity and amplifying effector T-cell function in the . This mechanism results in broader systemic immune activation compared to inhibitors targeting later-stage checkpoints, but it also contributes to increased immune-related toxicities, such as , , and endocrinopathies, with grade 3-4 adverse events occurring in up to 25-30% of patients on monotherapy. Response rates with CTLA-4 inhibitors as monotherapy are approximately 20%, yet these are often profound and durable in responders, yielding long-term survival benefits in a subset of patients. The pioneering CTLA-4 inhibitor, (Yervoy), was approved by the FDA in March 2011 as the first-in-class agent for unresectable or metastatic and is typically dosed at 1-3 mg/kg intravenously every 3 weeks for four cycles. In October 2022, tremelimumab (Imjudo) became the second approved CTLA-4 inhibitor, authorized in combination with for unresectable , featuring an initial priming dose of 300 mg followed by interval dosing. These approvals marked pivotal advancements in early blockade, with ipilimumab's development grounded in phase III trials demonstrating unprecedented survival improvements in advanced .

PD-1/PD-L1 Inhibitors

The PD-1/PD-L1 axis plays a critical role in immune regulation, where (PD-1), expressed on activated T cells, binds to (PD-L1) on tumor cells and antigen-presenting cells, leading to T-cell exhaustion, reduced proliferation, and induction of , thereby suppressing antitumor immunity. Inhibitors targeting this axis are monoclonal antibodies that block the PD-1/PD-L1 interaction, preventing inhibitory signaling and reinvigorating T-cell activity to enhance cytotoxic responses against tumors. Approved PD-1 inhibitors include nivolumab, a fully IgG4 first approved by the FDA in December 2014 for unresectable or metastatic ; pembrolizumab, a humanized IgG4 approved in September 2014 for advanced and later expanded to numerous indications; cemiplimab, approved in September 2018 for metastatic or locally advanced ; and dostarlimab, approved in April 2021 for mismatch repair-deficient recurrent or advanced . These agents broadly block PD-1 engagement with both and PD-L2 ligands, restoring T-cell function across PD-1-expressing immune cells. Approved PD-L1 inhibitors include , a IgG1 first approved by the FDA in May 2016 for locally advanced or metastatic urothelial carcinoma; , a IgG1 approved in March 2017 for metastatic ; and , a IgG1 approved in May 2017 for locally advanced or metastatic urothelial carcinoma. Unlike PD-1 inhibitors, these target only , sparing interactions with PD-L2, which may result in a potentially lower incidence of certain immune-related adverse events, such as endocrine toxicities, due to preserved PD-1/PD-L2 signaling in non-tumor tissues.

Other Cell Surface Inhibitors

Beyond the well-established CTLA-4 and PD-1/PD-L1 pathways, other cell surface immune checkpoints have emerged as promising targets for , particularly those expressed on T cells and other immune effectors within the . These include lymphocyte activation gene-3 (LAG-3), T-cell immunoreceptor with Ig and ITIM domains (), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), and V-domain Ig-containing suppressor of T-cell activation (), each modulating T-cell exhaustion and suppression through distinct ligand interactions. LAG-3 is an inhibitory receptor primarily expressed on activated CD4+ and CD8+ T cells, natural killer cells, and regulatory T cells, where it binds to class II molecules on antigen-presenting cells, thereby dampening T-cell proliferation and production while synergizing with PD-1 to exacerbate T-cell exhaustion. This cooperative suppression limits antitumor immunity, making LAG-3 blockade a strategic addition to PD-1 inhibition. In 2022, the U.S. (FDA) approved , a targeting LAG-3, in fixed-dose combination with nivolumab (Opdualag) for adult and pediatric patients (aged 12 years and older) with unresectable or metastatic , based on phase 3 trial data showing improved compared to nivolumab monotherapy. As of 2025, ongoing phase 3 trials continue to evaluate LAG-3 inhibitors in combination regimens for non-small cell (NSCLC), with preliminary data indicating potential expansions in approved indications, though no new FDA approvals for NSCLC combinations have been granted to date. TIGIT functions as an inhibitory receptor on T cells and natural killer cells, featuring immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domains that bind poliovirus receptor (PVR) ligands such as and CD112, thereby competing with the costimulatory receptor CD226 (DNAM-1) to suppress cytotoxic activity and promote regulatory T-cell function. This antagonism inhibits immune synapse formation and T-cell activation in the . Tiragolumab, an anti- , advanced to phase 3 trials, particularly in NSCLC, where combinations with (anti-PD-L1) showed promising early results; for instance, the phase 2 trial in PD-L1-positive NSCLC demonstrated an objective response rate of 37% for the tiragolumab- combination versus 21% for alone. The phase 3 SKYSCRAPER-02 trial reported enhanced ( of 0.70) when added to plus versus alone in extensive-stage . However, multiple phase 3 trials failed to meet endpoints, including SKYSCRAPER-01 in NSCLC (April 2025), SKYSCRAPER-07 in esophageal (October 2025), and a study, leading to discontinue development of tiragolumab and the broader program in July 2025 after enrolling over 5,000 patients. TIM-3, expressed on exhausted T cells, dendritic cells, and myeloid cells, exerts inhibitory effects by binding galectin-9, a soluble that promotes T-cell and impairs Th1 responses, thereby fostering an immunosuppressive tumor environment. While preclinical models demonstrate that TIM-3 blockade enhances antitumor immunity, particularly in combination with PD-1 inhibitors, clinical development remains in early phases, with phase 1 trials ongoing as of 2025 showing modest monotherapy activity but synergistic potential in solid tumors. VISTA, a B7 family member expressed on myeloid cells and T cells, inhibits T-cell activation through interactions with PSGL-1 and VSIG-3, with binding affinity heightened in the acidic tumor microenvironment (pH ~6.5-7.0) due to protonation of key histidine residues, thereby suppressing innate and adaptive immunity in hypoxic, glycolytic tumors. Preclinical studies highlight VISTA's role in myeloid-derived suppressor cell-mediated evasion, and pH-selective antibodies like SNS-101 are in early clinical testing, aiming to selectively block this pathway without systemic effects.

Intracellular Checkpoint Inhibitors

Intracellular checkpoint inhibitors target signaling pathways within immune cells, particularly T cells, that dampen activation and effector functions downstream of cell surface receptors. Unlike surface-targeted therapies, these inhibitors address post-receptor brakes, often requiring gene editing or small molecules to modulate intracellular proteins. A prominent example is the cytokine-inducible SH2-containing protein (), a negative regulator of T-cell responses. CISH inhibits Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling following stimulation by cytokines such as interleukin-2 (IL-2) and IL-15, thereby limiting T-cell , , and metabolic . This suppression occurs by recruiting phosphatases to dephosphorylate STAT proteins, curtailing downstream essential for effector . Genetic of CISH via CRISPR-Cas9 enhances T-cell sensitivity to these s, boosting JAK-STAT activation, mTOR signaling, and production, which collectively amplify anti-tumor activity in preclinical models. Preclinical studies using CRISPR-mediated CISH knockout in the 2020s have demonstrated significantly enhanced T-cell persistence and anti-tumor efficacy, with engineered cells showing prolonged survival and increased infiltration into solid tumors compared to wild-type counterparts. These findings have advanced to early clinical testing, including phase 1 trials initiated in 2024-2025 evaluating CISH-knockout tumor-infiltrating lymphocytes (TILs) or chimeric antigen receptor (CAR) T cells for augmentation in solid tumors, such as metastatic gastrointestinal cancers, where initial data indicate improved safety and preliminary anti-tumor responses. Other intracellular targets include non-receptor type 2 (PTPN2), a phosphatase that attenuates (TCR) and signaling by dephosphorylating key kinases like JAK1/2 and ZAP70. Small molecule inhibitors of PTPN2, such as ABBV-CLS-484, have entered phase 1 trials by 2025, showing potential to enhance T-cell activation and tumor responsiveness in immunotherapy-resistant settings. Additionally, the transcription factor TOX drives T-cell exhaustion by enforcing epigenetic changes that stabilize dysfunctional states during chronic exposure; emerging epigenetic modulators, including , are being explored to disrupt TOX-mediated and reinvigorate exhausted T cells. These intracellular approaches offer advantages over traditional intravenous antibody-based therapies, including the potential for oral delivery that improves patient convenience, tissue penetration, and cost-effectiveness. Furthermore, they exhibit synergy with cell surface checkpoint blockers, as CISH inhibition reduces PD-1 expression on T cells, potentially broadening responses in combination regimens.

Clinical Use

Approved Indications

Checkpoint inhibitors have received regulatory approvals from the U.S. Food and Drug Administration (FDA) and the (EMA) for treating various advanced or metastatic solid tumors, primarily as monotherapy or in combination regimens for patients who have progressed on prior therapies or meet specific criteria. In melanoma, ipilimumab (a CTLA-4 inhibitor) was the first checkpoint inhibitor approved by the FDA in 2011 as monotherapy for unresectable or metastatic disease, with subsequent approvals for pembrolizumab (a PD-1 inhibitor) in 2014 and nivolumab in 2014, both demonstrating superior overall survival compared to standard . First-line combination therapy with ipilimumab plus nivolumab or pembrolizumab has been approved since 2015 for advanced melanoma, achieving a 5-year overall of approximately 50% in clinical trials, representing a substantial improvement over historical rates of less than 20%. For non-small cell lung cancer (NSCLC), nivolumab received FDA approval in 2015 for squamous and non-squamous subtypes after platinum-based chemotherapy failure, with objective response rates (ORR) of 20% in second-line settings. Durvalumab (a PD-L1 inhibitor) was approved in 2018 for unresectable stage III NSCLC following chemoradiotherapy, based on improved progression-free survival. Pembrolizumab and atezolizumab are approved as first-line monotherapy or with chemotherapy for metastatic NSCLC with PD-L1 expression, particularly in subsets with microsatellite instability-high (MSI-H) tumors where ORR exceeds 50%. Additional approvals include for locally advanced or metastatic urothelial carcinoma in cisplatin-ineligible patients since 2016, nivolumab for advanced in combination with since 2018, and for recurrent or metastatic head and neck with expression since 2016. In 2022, the FDA approved nivolumab plus as first-line therapy for unresectable advanced or metastatic esophageal . Patient selection often relies on biomarkers such as tumor proportion score (TPS) ≥1% for NSCLC approvals of monotherapy, which correlates with higher response rates. MSI-H or mismatch repair-deficient (dMMR) status supports pan-tumor approvals, including for and across solid tumors since 2017, with enhanced efficacy in these subsets. In April 2025, the FDA approved nivolumab plus as first-line treatment for adults and pediatric patients (aged 12 years and older) with MSI-H or dMMR unresectable or metastatic .

Combination Therapies

Combination therapies involving checkpoint inhibitors aim to enhance antitumor immune responses by targeting multiple immunosuppressive pathways or integrating with other treatment modalities, thereby addressing mechanisms of resistance observed in monotherapy settings. The rationale for combining checkpoint inhibitors, such as CTLA-4 and PD-1 blockers, lies in their complementary mechanisms: CTLA-4 inhibition primarily acts early in T-cell priming to expand the T-cell repertoire and deplete regulatory T cells in lymphoid tissues, while PD-1 blockade functions later in the to reinvigorate exhausted effector T cells. This sequential blockade approach has demonstrated synergistic effects, leading to improved efficacy metrics like objective response rates (ORR) boosted by 10-20% in various trials, though it is associated with higher rates of immune-related adverse events (irAEs) due to amplified immune activation. Prominent examples of checkpoint inhibitor combinations include the pairing of nivolumab (anti-PD-1) with (anti-CTLA-4), evaluated in the phase 3 CheckMate 067 trial for advanced , where the combination yielded a 5-year overall (OS) rate of 52%, compared to 44% with nivolumab alone and 26% with ipilimumab monotherapy. Similarly, the RELATIVITY-047 trial in 2022 assessed (anti-LAG-3) plus nivolumab in untreated advanced , achieving a median (PFS) of 10.1 months versus 4.6 months with nivolumab alone, highlighting the benefit of dual blockade beyond PD-1/CTLA-4 axes. These regimens underscore how combining inhibitors of distinct checkpoints can overcome compensatory resistance pathways, though they necessitate careful toxicity management. Integrating checkpoint inhibitors with or further potentiates efficacy by leveraging treatment-induced immunogenic to increase presentation and T-cell infiltration. In the phase 3 KEYNOTE-189 trial, combined with pemetrexed-platinum in metastatic nonsquamous non-small cell (NSCLC) resulted in a median OS of 22.0 months, significantly outperforming alone (10.7 months). The PACIFIC trial demonstrated the value of sequential therapy, with consolidation following chemoradiotherapy in unresectable stage III NSCLC yielding a 5-year OS rate of 42.9%, versus 33.4% with , establishing this approach as a standard for consolidative . These combinations exploit the synergistic release of neoantigens and modulation of the immunosuppressive milieu to enhance checkpoint blockade. Emerging combinations in 2024-2025 trials continue to explore novel targets, such as PD-1/PD-L1 inhibitors with TIGIT blockers to disrupt additional inhibitory signaling in the tumor microenvironment. The phase 2 CITYSCAPE trial reported an ORR of 66% with tiragolumab (anti-TIGIT) plus atezolizumab (anti-PD-L1) in PD-L1-positive (TPS ≥50%) metastatic NSCLC, compared to 24% with atezolizumab alone, indicating potential for TIGIT as a synergistic target. Efforts to combine anti-CTLA-4 agents like ipilimumab with IDO1 inhibitors, such as epacadostat, have informed the field despite challenges; a phase 1/2 trial in unresectable melanoma showed modest activity with stable disease in 36% of patients but highlighted dose-limiting toxicities and limited durable responses, contributing to the broader understanding of metabolic checkpoint redundancies and the need for refined patient selection. These developments emphasize ongoing refinements in combination strategies to balance efficacy gains against toxicity profiles.

Adverse Effects

Checkpoint inhibitors, by unleashing T-cell activity against tumors, can also provoke immune overactivation leading to immune-related adverse events (irAEs), which affect various organs through autoimmune-like responses. These events occur due to nonspecific immune enhancement and are distinct from traditional toxicities, often requiring multidisciplinary management. Incidence varies by inhibitor type, with therapies increasing and severity. Dermatologic irAEs are the most frequent, manifesting as or pruritus in 40-50% of patients treated with PD-1/PD- inhibitors, with 3-4 severity in less than 5% of cases. These typically arise early in treatment and involve T-cell infiltration into the skin, resembling eczematous or maculopapular eruptions. , an autoimmune depigmentation, is also noted, particularly with PD-1 blockade. Gastrointestinal irAEs, such as and , affect up to 30% of patients on CTLA-4 inhibitors like , compared to 5-10% with PD-1 inhibitors. These result from mucosal driven by cytotoxic T cells and release, potentially leading to in severe cases. The gut plays a key role, with enrichment of conferring protection against by promoting regulatory T-cell responses and reducing . Endocrine irAEs include or in 10-15% of PD-1/ inhibitor recipients, often due to antithyroid antibodies, while occurs in 5-10% of CTLA-4 inhibitor users, causing pituitary and deficiencies. These events stem from immune attack on endocrine glands and may require lifelong replacement. Other irAEs encompass (3-5% incidence, predominantly with PD-1/ inhibitors and patients), characterized by interstitial ; (1-10%, elevated with combinations), involving hepatocellular injury; and rare neurologic events like (<1%), which can be life-threatening due to central nervous system infiltration. As of 2025, emerging insights highlight gut modulation via fecal transplantation, which has shown efficacy in reducing severity in cases, with clinical improvement in approximately 80% of patients and complete remission in over 50%.

Prevention and Management

Prevention of immune-related adverse events (irAEs) associated with checkpoint inhibitors involves and high suspicion for symptoms, but prophylactic agents such as corticosteroids are not routinely recommended. Emerging research also explores optimization prior to treatment, including the use of or fecal microbiota transplantation to modulate and potentially reduce gastrointestinal irAEs, though these approaches remain investigational. Monitoring for irAEs begins with baseline laboratory assessments, including and liver enzymes, to establish a reference for potential changes. Ongoing surveillance typically includes monthly checks of relevant labs and clinical evaluations, with severity graded using the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 to guide interventions. This systematic approach allows for early detection and timely management of toxicities. Management of irAEs prioritizes corticosteroids as first-line therapy, with administered at 1 mg/kg daily for grade 2 or higher events, tapered once symptoms improve. For refractory , (5 mg/kg) is recommended after failure of steroids, often leading to resolution in most cases. Endocrinopathies, such as or , are typically managed with lifelong , such as or , without necessitating discontinuation if controlled. Discontinuation of checkpoint inhibitors is advised permanently for grade 4 toxicities or recurrent grade 3 events to prevent life-threatening complications. Rechallenge with the same or alternative agents may be considered after resolution to grade 1 or lower, but only with close monitoring due to the risk of recurrence. Recent NCCN guideline updates (Version 2.2024) underscore the importance of multidisciplinary teams, involving oncologists, endocrinologists, and dermatologists, to coordinate care and optimize outcomes. is also emphasized, including provision of symptom recognition tools and emergency contact protocols to empower early reporting of potential irAEs.

History and Development

Discovery of Key Pathways

The discovery of CTLA-4 as a key immune regulatory molecule began with its identification in the late , but its role as an inhibitory checkpoint was elucidated through genetic and functional studies in the mid-1990s. In 1995, researchers generated CTLA-4-deficient mice, which rapidly developed fatal and multi-organ , demonstrating that CTLA-4 acts as a critical negative regulator of T-cell activation to prevent excessive immune responses. This finding highlighted CTLA-4's function in maintaining immune by competing with the co-stimulatory receptor for binding to B7 ligands on antigen-presenting cells, thereby dampening T-cell and production. Building on this, James Allison's laboratory in 1996 tested monoclonal antibodies against CTLA-4 in mouse models of cancer, showing that blockade of CTLA-4 enhanced T-cell-mediated tumor rejection, providing early evidence that interrupting this pathway could unleash antitumor immunity without solely relying on genetic knockout. Parallel efforts uncovered the PD-1 pathway, with Tasuku Honjo's group identifying PD-1 in 1992 as a novel member upregulated during in T-cell hybridomas, initially suggesting a role in regulation. Subsequent work by Honjo's team in 1999 identified (initially termed B7-H1) as a ligand for PD-1, expressed on various immune and non-immune cells, which delivers inhibitory signals upon binding to suppress T-cell activity. By 2002, studies linked PD-L1 expression on tumor cells to immune evasion, as tumors exploited this pathway to inhibit cytotoxic T-cell responses, marking a pivotal connection between PD-1/PD-L1 signaling and cancer progression. Early functional validation came in 1999-2000, when PD-1 mice exhibited mild autoimmune phenotypes, reinforcing its inhibitory role akin to CTLA-4 but with distinct tissue-specific effects. These discoveries culminated in preclinical demonstrations of therapeutic potential. In 1996, Leach and colleagues in Allison's lab reported that anti-CTLA-4 antibodies induced tumor in mice bearing immunogenic tumors, establishing CTLA-4 as a strategy to potentiate antitumor immunity. Similarly, Honjo's group in 2005 showed that PD-1 , using antibodies in mouse models, inhibited the hematogenous spread of poorly immunogenic tumors like by enhancing + T-cell recruitment and activation. Published primarily in high-impact journals such as and , these foundational studies shifted the paradigm in the early toward viewing CTLA-4 and PD-1 as molecular "brakes" on T cells, inspiring targeted immunotherapies to release these inhibitions for .

Milestones and Approvals

The development of checkpoint inhibitors reached a pivotal with the FDA's approval of , the first CTLA-4 inhibitor, on March 25, 2011, for the treatment of unresectable or metastatic in previously treated adults. This approval was based on results from the phase 3 CA184-024 trial, which demonstrated a median overall survival of 10.1 months with ipilimumab compared to 6.4 months with the gp100 vaccine alone ( 0.66; 95% CI, 0.51 to 0.87). Ipilimumab's approval marked the advent of immune checkpoint blockade in clinical practice, establishing a new paradigm for . The PD-1 inhibitor era began in 2014 with FDA approvals for nivolumab on December 22 for BRAF V600 wild-type unresectable or metastatic melanoma post-ipilimumab, and for pembrolizumab on September 4 for ipilimumab-refractory advanced melanoma. These were followed by expansions to non-small cell lung cancer (NSCLC) in 2015, with nivolumab approved on March 4 for squamous NSCLC after platinum-based chemotherapy and on October 9 for non-squamous NSCLC, and pembrolizumab on October 2 for PD-L1-positive NSCLC post-platinum therapy. The field expanded rapidly from 2016 to 2018 with PD-L1 inhibitors, including atezolizumab approved on May 18, 2016, for urothelial carcinoma, durvalumab on May 1, 2017, for urothelial carcinoma, and avelumab on March 23, 2017, for Merkel cell carcinoma, reflecting accelerated adoption across multiple tumor types. Subsequent years saw further innovations in combinations and novel targets. In 2022, the FDA approved tremelimumab in combination with on October 21 for unresectable , based on the phase 3 HIMALAYA trial showing a 22% reduction in death risk versus . Also in 2022, on March 18, nivolumab plus —the first LAG-3 inhibitor combination—was approved for unresectable or metastatic in adults and children aged 12 and older, demonstrating superior over nivolumab monotherapy in the RELATIVITY-047 trial. Additionally, on May 27, 2022, nivolumab (Opdivo) plus (Yervoy) received approval for first-line treatment of advanced or metastatic esophageal , supported by the phase 3 648 trial. In 2025, the FDA approved nivolumab plus on April 11 for unresectable or metastatic in adults who received prior . The foundational contributions to checkpoint inhibition were recognized with the 2018 in Physiology or Medicine awarded to and for their discoveries of CTLA-4 and PD-1 as cancer therapy targets. Approvals by the () have closely paralleled those of the FDA, with authorized in 2011, nivolumab and in 2015, and subsequent expansions aligning with U.S. indications for , NSCLC, and other cancers.

Current Research and Future Directions

New Targets and Therapies

Several phase 3 trials of investigational inhibitors, such as vibostolimab in fixed-dose combination with , for non-small cell lung cancer (NSCLC) were discontinued in 2024 (e.g., KEYVIBE-003, -007, -008) due to lack of efficacy, adverse events, or futility, with no FDA approvals as of November 2025. Earlier studies had demonstrated overall response rates (ORR) approaching 40% when combined with standard therapies, including enhanced efficacy in PD-L1-positive metastatic NSCLC from the KEYVIBE program. Roche's tiragolumab also failed a phase 3 trial in November 2024, highlighting challenges in development. Other emerging targets include TIM-3, with cobolimab showing promising activity in phase 2 trials reported in 2024, including acceptable safety and response rates in advanced NSCLC when combined with ; however, a phase 3 trial failed in July 2025. For ILT4 inhibition, monalizumab is being investigated in trials, though phase 3 studies like INTERLINK-1 were discontinued in 2022 due to futility, prompting ongoing exploration in combination regimens. Bispecific antibodies targeting PD-1 and LAG-3, such as tebotelimab and RO7247669, have demonstrated antitumor activity in early-phase (phase 1) trials by simultaneously blocking these checkpoints. Novel modalities extend beyond traditional monoclonal antibodies to intracellular targets, including CRISPR-Cas9-mediated knockout of CISH in (TILs) or primary T-cells entering early clinical trials in 2025, which aim to enhance T-cell function by disrupting signaling checkpoints independently of expression. Such gene editing approaches have shown potential to boost effector function and overcome resistance in preclinical and early clinical studies, including enhanced serial killing in cells. In 2025 advances, (TCM)-PD-1 combinations, such as Chinese yam polysaccharide with anti-PD-1 agents, have shown enhanced antitumor effects in preclinical models of by modulating and enhancing T-cell responses. Neoadjuvant immune checkpoint inhibitors (ICIs) combined with in have achieved pathological complete response rates of approximately 25% in hormone receptor-positive tumors, supporting their role in early-stage disease management.

Challenges and Biomarkers

Despite their transformative impact on cancer treatment, checkpoint inhibitors face significant challenges in efficacy, with primary resistance affecting 60-70% of patients who do not respond to therapy, often due to immunosuppressive tumor microenvironments or low tumor immunogenicity. Acquired resistance further complicates outcomes, emerging in initially responsive patients through dynamic changes in the tumor microenvironment (TME), such as upregulation of transforming growth factor-β (TGF-β), which promotes fibrosis, limits T-cell infiltration, and fosters regulatory T-cell expansion. These mechanisms highlight the need for adaptive resistance models that account for evolving immune evasion, including compensatory upregulation of alternative checkpoints like TIM-3 following PD-1 blockade. Biomarkers play a crucial role in patient stratification, though many have limitations in predictive accuracy. (IHC) assessment of expression offers limited predictive value for checkpoint inhibitor response, with efficacy observed in PD-L1-negative tumors and inconsistent correlation across assays and tumor types, contributing to its endorsement in only about 29% of U.S. approvals. In contrast, (TMB), defined as greater than 10 mutations per megabase, correlates with improved objective response rates (ORR) of approximately 40% in non-small cell and other solid tumors, reflecting higher neoantigen load and immune recognition. instability-high (MSI-H) status predicts near-universal and durable responses across tumor types, leading to tissue-agnostic approval of for MSI-H/dMMR cancers due to robust T-cell infiltration and neoantigen diversity. As of 2025, emerging biomarkers include gut microbiome profiling, where enrichment of has been associated with enhanced response to PD-1 blockade in advanced non-small cell lung cancer, potentially through improved maturation and T-cell priming. (ctDNA) monitoring provides dynamic insights, with early ctDNA clearance post-therapy predicting prolonged survival and response in metastatic and urothelial treated with checkpoint inhibitors. These tools underscore the push toward multimodal stratification to overcome resistance. Addressing these challenges requires combination therapies to achieve a 20-30% efficacy boost over monotherapy, targeting adaptive resistance pathways like TGF-β signaling or to expand the responder population beyond the current 20-40% benchmark.

References

  1. [1]
    Immune Checkpoint Inhibitors and Their Side Effects
    Jul 7, 2025 · Immune checkpoint inhibitors block the signals from these checkpoints, allowing the immune system to find and attack the cancer cells. On this ...Missing: definition | Show results with:definition
  2. [2]
    Mechanisms of immune checkpoint inhibitors - Nature
    Jan 4, 2024 · Cancer immunotherapy with ICIs by inhibition of negative immune regulation to enhance immune activity against cancer cells, which won the 2018 ...
  3. [3]
    Checkpoint inhibitors | Types of immunotherapy | Cancer Research UK
    Checkpoint inhibitors are a type of immunotherapy. These drugs block different proteins called checkpoint proteins that stop the immune system.
  4. [4]
    Immune Checkpoint Inhibitors in Cancer Therapy - PMC
    ICIs are cancer immunotherapies that boost anti-cancer immune responses by targeting immunologic receptors on the surface of T-lymphocytes (Table 1) [13].
  5. [5]
    Recent developments on checkpoint inhibitors, CAR T cells, and ...
    Mar 28, 2025 · While, cytokine therapy is accompanied by the use of cytokines such as interleukin-2 (IL-2) to stimulate immune cell activity and proliferation, ...
  6. [6]
    A decade of immune-checkpoint inhibitors in cancer therapy - Nature
    Jul 30, 2020 · Immunotherapy using immune-checkpoint modulators revolutionizes the oncology field far beyond their remarkable clinical efficacy in some patients.
  7. [7]
    Immune checkpoint inhibitors: breakthroughs in cancer treatment
    May 24, 2024 · The discovery and development of immune checkpoint inhibitors (ICIs) substantially enhanced tumor treatment outcomes.
  8. [8]
    Lessons learned from a decade of immune checkpoint inhibition - NIH
    Apr 4, 2025 · ICIs have transformed the treatment of numerous cancer types and, remarkably, some patients with end-stage metastatic disease can achieve durable, complete ...
  9. [9]
    Resistance mechanisms to immune checkpoint inhibitors
    Jan 15, 2025 · The durability of responses translated into long-term survival benefits is considered a significant hallmark of cancer immunotherapy. In 2011, ...
  10. [10]
    How many people in the US are eligible for and respond to ...
    Jan 30, 2025 · Eleven ICI drugs have been approved for 20 tumor types in the metastatic setting. The estimated eligibility for ICIs increased from 1.54% in ...
  11. [11]
    PD-1 and PD-L1 Inhibitors Show Dramatic Response Rates in NSCLC
    Patients with high levels of PD-L1 expression had an ORR of nearly 70%, while patients with lower rates of expression experienced a lower response rate, the ...
  12. [12]
    Development of PD-1 and PD-L1 inhibitors as a form of cancer ...
    Jan 23, 2018 · The ORR was 45.2% in the patients with PD-L1 ≥ 50%, including 43.9% in previously-treated patients and 50.0% in untreated patients. These values ...Melanoma · Non-Small Cell Lung Cancer · Urothelial Cancer
  13. [13]
    Final, 10-Year Outcomes with Nivolumab plus Ipilimumab in ...
    May 15, 2025 · Among these patients, 10-year overall survival rates were 86% with nivolumab-plus-ipilimumab, 85% with nivolumab, and 79% with ipilimumab ( ...Missing: pre- | Show results with:pre-
  14. [14]
    Long-term Metastatic Melanoma Survival Dramatically Improves on ...
    Sep 15, 2024 · Half of patients with metastatic melanoma treated with a combination of immune checkpoint inhibitors survive cancer-free for 10 years or more.Missing: pre- | Show results with:pre-
  15. [15]
    Melanoma Treatments and Mortality Rate Trends in the US, 1975 to ...
    Dec 6, 2022 · However, with the use of novel therapies, the 5-year overall survival rate has increased from 5% to greater than 50%. A frequent cause of ...
  16. [16]
    Biomarkers for Response to Anti–PD-1/Anti–PD-L1 Immune ...
    May 17, 2023 · A large meta-analysis assessing the ability of various biomarkers to predict responses to immune checkpoint inhibition.
  17. [17]
    The Nobel Prize in Physiology or Medicine 2018 - NobelPrize.org
    The Nobel Prize in Physiology or Medicine 2018 was awarded jointly to James P. Allison and Tasuku Honjo for their discovery of cancer therapy.
  18. [18]
    A guide to cancer immunotherapy: from T cell basic science ... - Nature
    May 20, 2020 · CTLA4 restrains T cell activation through multiple mechanisms: by directly antagonizing CD28, by competing for co-stimulatory ligands, by ...
  19. [19]
    CTLA-4 and PD-1 Pathways - PubMed Central - NIH
    The CTLA-4 and PD-1 immune checkpoint pathways downregulate T-cell activation to maintain peripheral tolerance, and can be exploited by tumors to induce an ...
  20. [20]
    Regulatory mechanisms of PD-1/PD-L1 in cancers - Molecular Cancer
    May 18, 2024 · However, cancer cells exploit the PD-1/PD-L1 axis to cause immune escape in cancer development and progression. Blockade of PD-1/PD-L1 by ...
  21. [21]
    Current Understanding of Cytotoxic T Lymphocyte Antigen-4 (CTLA ...
    CTLA-4 negatively regulates T-cell activation and plays an important role in the suppressive function of regulatory T (Treg) cells (Krummel and Allison ...
  22. [22]
    Fundamental Mechanisms of Immune Checkpoint Blockade Therapy
    Immune checkpoint blockade inhibits T cell–negative costimulation in order to unleash antitumor T-cell responses that recognize tumor antigens. Importantly, the ...Abstract · Introduction · Mechanisms of Negative... · Mechanisms of Action of PD-1...
  23. [23]
    A Review of the Pharmacokinetic Characteristics of Immune ... - NIH
    Jan 20, 2023 · The linear and non-specific elimination pathways predominate following saturation of the target receptor, resulting in a half-life of three to ...
  24. [24]
    Ipilimumab: An Anti-CTLA-4 Antibody for Metastatic Melanoma - PMC
    Here, we review the mechanism of action, preclinical data, and multiple clinical trials that led to FDA approval of ipilimumab for metastatic melanoma.
  25. [25]
    Dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in ...
    Jul 27, 2024 · Furthermore, the study revealed that the combined blockade of PD-1 and CTLA-4 enhanced the infiltration of Teffs, thereby producing a highly ...
  26. [26]
    The future of targeting cytotoxic T-lymphocyte-associated protein-4
    Compared to PD1/PDL1 inhibition, CTLA-4 blockade faces two main hurdles: lower rate of responses and higher toxicity [60]. ... toxicities associated with a CTLA-4 ...
  27. [27]
    FDA Approves YERVOY™ (ipilimumab) for the Treatment of Patients ...
    Mar 25, 2011 · Blockade of CTLA-4 has been shown to augment T-cell activation and proliferation. The mechanism of action of ipilimumab's effect in patients ...
  28. [28]
    FDA approves tremelimumab in combination with durvalumab
    Oct 24, 2022 · FDA approves tremelimumab in combination with durvalumab for unresectable hepatocellular carcinoma. On October 21, 2022, the Food and Drug ...
  29. [29]
    PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer ... - NIH
    Aug 23, 2017 · Atezolizumab is an FDA approved the drug for urothelial ... PD-1 inhibitors are being treated in combination with antineoplastic agents ...
  30. [30]
    [PDF] 125554Orig1s000 - accessdata.fda.gov
    Dec 22, 2014 · July 30, 2014, submitted under section 351 of the Public Health Service Act for Opdivo. (nivolumab) Injection for Intravenous Infusion. We ...
  31. [31]
    Keytruda (Pembrolizumab): First PD-1 Inhibitor Approved for ...
    11. Inman S. FDA approves pembrolizumab for advanced melanoma. OncLive. September 4, 2014. www.onclive.com/web-exclusives/FDA ...
  32. [32]
    FDA grants accelerated approval to dostarlimab-gxly for dMMR ...
    Apr 22, 2021 · On April 22, 2021, the Food and Drug Administration granted accelerated approval to dostarlimab-gxly (Jemperli, GlaxoSmithKline LLC) for adult ...
  33. [33]
    Atezolizumab for Urothelial Carcinoma - FDA
    May 19, 2016 · On May 18, 2016, the U. S. Food and Drug Administration gave accelerated approval to atezolizumab injection (Tecentriq, Genentech, Inc.)
  34. [34]
    Avelumab (BAVENCIO) - FDA
    May 30, 2017 · On March 23, 2017, the U.S. Food and Drug Administration granted accelerated approval to avelumab (BAVENCIO, EMD Serono, Inc.) for the treatment ...
  35. [35]
    Durvalumab (Imfinzi) - FDA
    May 1, 2017 · On May 1, 2017, the U.S. Food and Drug Administration granted accelerated approval to durvalumab (IMFINZI, AstraZeneca UK Limited) for the ...
  36. [36]
    UPDATES IN ENDOCRINE IMMUNE-RELATED ADVERSE ...
    Theoretically, autoimmune toxicity of anti-PD-L1 antibodies is less intense than anti-PD-1 agents, taking into consideration that PD-1/PD-L2 activity is still ...
  37. [37]
    Targeting LAG-3, TIM-3, and TIGIT for cancer immunotherapy
    Sep 5, 2023 · Tim-3/galectin-9 pathway: regulation of Th1 immunity through promotion of CD11b+Ly-6G+ myeloid cells. J Immunol. 2010;185(3):1383–92. CAS ...Anti-Lag-3 Mabs · Anti-Tigit Mabs · Lag-3-Pd-1/pd-L1/ctla-4/tigit...<|separator|>
  38. [38]
    Relatlimab: a novel drug targeting immune checkpoint LAG-3 in ...
    On 18 March 2022, the U.S. FDA approved the fixed-dose combination of relatlimab and nivolumab for the treatment of unresectable or metastatic melanoma in adult ...Mechanism of action and... · Relatlimab, the first LAG-3... · Results after 2 years of...
  39. [39]
    FDA approves Opdualag for unresectable or metastatic melanoma
    Mar 21, 2022 · Opdualag is a fixed-dose combination of the LAG-3-blocking antibody relatlimab and the programmed death receptor-1 blocking antibody nivolumab.
  40. [40]
    New Therapeutic Targets TIGIT, LAG-3 and TIM-3 in the Treatment ...
    Unfortunately, the NCCN guidelines for the treatment of non-small-cell lung cancer from January 2025 do not indicate the place of drugs using the blockade of ...
  41. [41]
    Tiragolumab and TIGIT: pioneering the next era of cancer ... - NIH
    Jun 11, 2025 · Phase II and III trials, including the CITYSCAPE trial for non-small cell lung cancer (NSCLC), have shown improved objective response rates (37% ...
  42. [42]
    SKYSCRAPER-02: Tiragolumab in Combination With Atezolizumab ...
    Nov 17, 2023 · The combination was well tolerated and showed clinically meaningful improvements in antitumor response, PFS, and OS versus atezolizumab alone, ...
  43. [43]
    Promising New Anti‐TIGIT Agents: Stealthy Allies in Cancer ...
    Apr 22, 2025 · Enhanced Treatment Outcomes: TIGIT inhibitors and other checkpoint inhibitors increase overall response rates (ORR) and disease control rates ( ...
  44. [44]
    The TIM3/Gal9 Signaling Pathway: An Emerging Target for Cancer ...
    The interaction of TIM3 and Galectin 9 (Gal9) suppresses anti-tumor immunity mediated by innate as well as adaptive immune cells.
  45. [45]
    First-in-human phase I open-label study of the anti–TIM-3 ...
    Jul 9, 2025 · Studies in preclinical mouse tumor models show that although TIM-3 inhibitors have modest antitumor activity as monotherapy, they demonstrate ...
  46. [46]
    VISTA checkpoint inhibition by pH-selective antibody SNS-101 with ...
    Apr 4, 2024 · This interaction is particularly relevant in the acidic tumor microenvironment (TME), characterized by extracellular acidosis due to deregulated ...
  47. [47]
    A highly potent anti-VISTA antibody KVA12123 - a new immune ...
    Dec 11, 2023 · The pH in the tumor microenvironment can vary from neutral pH 7.4 to more acidic pH 5.5, depending on the level of hypoxia and glycolysis (29–31) ...
  48. [48]
    Targeting a cytokine checkpoint enhances the fitness of armored ...
    Feb 4, 2021 · This gain of effector function is attributed to enhanced IL-15 signaling secondary to CISH KO, with consequent activation of the Akt/mTORC1/c- ...
  49. [49]
    Disruption of CISH promotes the antitumor activity of human T cells ...
    In this study, we ablated CISH in T cells with CRISPR-Cas9 and found that the sensitivity of T cells to TCR and cytokine stimulation was increased. In addition, ...Missing: 2020s | Show results with:2020s
  50. [50]
    A small molecule inhibitor of PTP1B and PTPN2 enhances T cell ...
    Jul 27, 2023 · The inhibition of protein tyrosine phosphatases 1B (PTP1B) and N2 (PTPN2) has emerged as an exciting approach for bolstering T cell ...
  51. [51]
    TOX transcriptionally and epigenetically programs CD8+ T cell ...
    Here we identify the HMG-box transcription factor TOX as a central regulator of T ex cells in mice. TOX is largely dispensable for the formation of T eff and T ...
  52. [52]
    Small Molecule Agents Targeting PD-1 Checkpoint Pathway for ...
    May 1, 2022 · Table 1 Advantages of small molecule agents over antibodies to target PD-1 and other immune checkpoint pathways.<|separator|>
  53. [53]
    FDA Approval Timeline of Active Immunotherapies | CRI
    Checkpoint inhibitors, in particular PD-1 and PD-L1, have been integrated into standard-of-care across many cancer types. The CRI Anna-Maria Kellen Clinical ...
  54. [54]
    Oncology (Cancer)/Hematologic Malignancies Approval Notifications
    On September 19, 2025, the Food and Drug Administration approved pembrolizumab and berahyaluronidase alfa-pmph (Keytruda Qlex, Merck) for subcutaneous injection ...FDA approves darolutamide... · FDA grants accelerated... · FDA approves belzutifan
  55. [55]
    Trends in complexity of single-agent and combination therapies for ...
    Nov 15, 2024 · Among the 292 approval entries and 110 drugs, 193 (66.1%) were monotherapies and 99 (33.9%) were combinations. Combinations, when compared with ...
  56. [56]
    The efficacy and safety of combination of PD-1 and CTLA-4 inhibitors
    Oct 25, 2019 · Combination treatment of α-PD-1 and α-CTLA-4 is a feasible strategy with enhanced efficacy and acceptable adverse event.
  57. [57]
    Immune checkpoint inhibitor combinations in solid tumors - NIH
    The goal of checkpoint inhibitor combination therapy is to improve clinical response and minimize toxicities.Immune Checkpoint Molecules... · Ctla-4 Monoclonal Antibodies... · Priming T-Cell Response
  58. [58]
    Five-Year Survival with Combined Nivolumab and Ipilimumab in ...
    Sep 27, 2019 · Overall survival at 5 years was 52% in the nivolumab-plus-ipilimumab group and 44% in the nivolumab group, as compared with 26% in the ipilimumab group.
  59. [59]
    Relatlimab and Nivolumab versus Nivolumab in Untreated ...
    Jan 5, 2022 · RELATIVITY-047 is a phase 3 trial that evaluated the dual inhibition of LAG-3 and PD-1 using a new combination of relatlimab, a human IgG4 ...
  60. [60]
    Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell ...
    Apr 16, 2018 · The addition of pembrolizumab to chemotherapy resulted in significantly higher rates of response and longer progression-free survival than chemotherapy alone ...
  61. [61]
    Five-Year Survival Outcomes From the PACIFIC Trial: Durvalumab ...
    Feb 2, 2022 · An estimated 42.9% of patients randomly assigned to durvalumab remain alive at 5 years and 33.1% of patients randomly assigned to durvalumab ...
  62. [62]
    Primary analysis of a randomized, double-blind, phase II study of the ...
    May 25, 2020 · We conducted this phase II trial to confirm the efficacy and safety of tira plus atezo (TA) compared to placebo plus atezo (PA) in 1L NSCLC.
  63. [63]
    Phase 1/2 study of epacadostat in combination with ipilimumab in ...
    Mar 20, 2019 · During the conduct of this trial, the emergence and clinical success of PD-1 inhibitors led to a shift in the melanoma treatment landscape.
  64. [64]
    Immune-related adverse events of immune checkpoint inhibitors - NIH
    May 25, 2023 · We summarize and discuss the mechanisms of action of immune checkpoint inhibitors, the different types of immune-related adverse events and their possible ...
  65. [65]
    Focus on Immune Checkpoint Inhibitors-related Intestinal Inflammation
    In contrast, increased Bacteroides fragilis, due to its anti-inflammatory activity, is considered a protective factor against ICI colitis. In particular, CTLA- ...
  66. [66]
    Microbiome alteration via fecal microbiota transplantation is effective ...
    Jan 2, 2024 · Microbiome alteration via fecal microbiota transplantation is effective for refractory immune checkpoint inhibitor–induced colitis.
  67. [67]
    Management of Immune-Related Adverse Events in Patients ...
    ICPi therapy should be continued with close monitoring for grade 1 toxicities, except for some neurologic, hematologic, and cardiac toxicities.
  68. [68]
    Achilles' Heel of currently approved immune checkpoint inhibitors
    This review aims to present the pathogenesis, clinical manifestations, incidence, and treatment strategies of various irAEs through 49 clinical trials from ...
  69. [69]
    Management of Immunotherapy-Related Toxicities - Guidelines Detail
    Guidelines for Patients ; Immunotherapy Side Effects: CAR T-Cell Therapy-English Version 2024 ; Immunotherapy Side Effects: Immune Checkpoint Inhibitors-English ...
  70. [70]
    Immune checkpoint inhibitor rechallenge after immune-related ... - NIH
    Dec 3, 2024 · In this updated, largest cohort study on rechallenge (NCT04696250), we observed a 31.8% recurrence rate of the same irAE postrechallenge with ...
  71. [71]
    Improved Survival with Ipilimumab in Patients with Metastatic ...
    Jun 5, 2010 · This phase 3 study showed that ipilimumab, either alone or with gp100, improved overall survival as compared with gp100 alone in patients with ...
  72. [72]
    FDA approves Opdivo in combination with chemotherapy and ...
    May 31, 2022 · FDA approves Opdivo in combination with chemotherapy and Opdivo in combination with Yervoy for first-line esophageal squamous cell carcinoma ...
  73. [73]
    Introducing CRI's 2025 Cancer Immunotherapy Insights + Impact ...
    Jun 4, 2025 · Immune checkpoint inhibitors remain the most widely used class, accounting for 81% of total approvals. In 2024, we also saw the first approvals ...
  74. [74]
    P14.01 Phase 3 Study of First-Line Pembrolizumab ± Vibostolimab ...
    Jun 27, 2025 · P14.01 Phase 3 Study of First-Line Pembrolizumab ± Vibostolimab (anti-TIGIT) in Patients With PD-L1‒Positive Metastatic NSCLC.
  75. [75]
    Cobolimab/Dostarlimab Combination Shows Initial Efficacy and ...
    Jan 10, 2024 · Cobolimab plus dostarlimab-gxly showed activity and acceptable safety across all dose levels assessed in patients with non–small cell lung cancer.
  76. [76]
    INTERLINK-1 Trial Terminated Following Failed Futility Analysis of ...
    Aug 1, 2022 · The phase 3 INTERLINK-1 trial was discontinued after monalizumab plus cetuximab failed to meet a predefined threshold for efficacy compared with cetuximab ...Missing: ILT4 | Show results with:ILT4
  77. [77]
    Tebotelimab, a PD-1/LAG-3 bispecific antibody, in patients with ...
    Jan 24, 2023 · Tebotelimab, also known as MGD013, is a bispecific tetravalent DART molecule that can bind both PD-1 and LAG-3. We initiated an open-label, ...
  78. [78]
    the randomized phase 1b/2 Morpheus-Melanoma trial - Nature
    Sep 24, 2025 · In phase 2/3 trials, concurrent checkpoint inhibition of PD-1 and LAG-3 led to an improved progression-free survival versus PD-1 inhibition ...
  79. [79]
    First-in-Human Data Highlight Potential of Intracellular Checkpoint ...
    May 3, 2025 · The phase 1 study (NCT04426669) evaluated the effects of administering autologous neoantigen-reactive tumor-infiltrating lymphocytes (TILs) with ...
  80. [80]
    Targeting the intracellular immune checkpoint CISH with CRISPR ...
    Apr 29, 2025 · The aim of this study was to determine the safety and anti-tumour activity of knockout of CISH, which encodes cytokine-inducible SH2-containing ...
  81. [81]
  82. [82]
    Neoadjuvant Immune Checkpoint Inhibitors Plus Chemotherapy in ...
    Aug 29, 2024 · In HR+/ERBB2− tumors, the addition of ICI improved the overall pCR rate from 14.8% to 24.6% (Δ = 9.8%; OR, 1.87; 95% CI, 1.49-2.36).
  83. [83]
    Mechanisms of Resistance to Immune Checkpoint Blockade - NIH
    CTLA-4 binds to B7 on APCs, competitively disrupting the B7-CD28 “second-signal” which is needed for T-cell activation and proliferation, thus serving as a ...
  84. [84]
    Primary and Acquired Resistance to Immunotherapy with ...
    Feb 15, 2025 · In the tumor microenvironment TGF-b is a master regulator, with broad immunosuppressive effects on CD8+ T cells, blocking their infiltration and ...2.1. 3 Macrophage Polarity · 2.1. 4 Neutrophils · 3.1. 1 Pd-L1 And Tumor...
  85. [85]
    Primary, Adaptive and Acquired Resistance to Cancer Immunotherapy
    Primary and Adaptive Resistance to Immunotherapy. Patients who have primary resistance to checkpoint inhibitors do not respond to the initial therapy.
  86. [86]
    The role of PD-L1 expression as a predictive biomarker
    Oct 26, 2019 · The goal of our study was to evaluate PD-L1 as a predictive biomarker based on all US Food and Drug Administration (FDA) drug approvals of immune checkpoint ...
  87. [87]
    Tumor mutation burden predicts response and survival to immune ...
    In addition, CheckMate 568 found that for NSCLC patients treated with ICIs, the objective response rate (ORR) no longer increased when TMB ≥10 mut/Mb (40).
  88. [88]
    Immune checkpoint inhibitors for the treatment of MSI-H/MMR ... - NIH
    Oct 14, 2019 · Studies have shown a sustained clinical response to immune checkpoint inhibitors with dramatic clinical improvement in patients with MSI-H/MMR-D CRC.
  89. [89]
    The gut microbiome and cancer response to immune checkpoint ...
    Feb 3, 2025 · Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat Med ...Bacteria that promote... · Bacteria associated with... · Bacterial metabolites as...
  90. [90]
    Circulating tumor DNA monitoring and blood tumor mutational ...
    May 28, 2025 · This study evaluates changes in circulating tumor DNA (ctDNA) and blood tumor mutational burden (bTMB) for associations with response to programmed cell death ...
  91. [91]
    Predictive models for immune checkpoint inhibitor response in cancer
    Several approvals occurred after extensive validation from clinical trials. Currently, there are three FDA-approved predictive biomarkers namely PD-L1, MSI ...<|separator|>