A Comprehensive Clinical and Pharmacological Monograph on Recombinant Anti-PD-1 Humanized Monoclonal Antibodies: A Paradigm Shift in Modern Oncology
Executive Summary
The advent of recombinant anti-Programmed Death-1 (PD-1) humanized monoclonal antibodies represents one of the most significant advances in oncology of the 21st century. This therapeutic class has fundamentally altered the treatment landscape and prognosis for a multitude of advanced malignancies.[1] These agents function as immune checkpoint inhibitors, operating on a core principle of releasing the endogenous "brakes" on the immune system. By blocking the interaction between the PD-1 receptor on T-cells and its ligands, primarily Programmed Death-Ligand 1 (PD-L1), these therapies restore the immune system's intrinsic ability to recognize and eliminate cancer cells.[3]
This monograph provides a comprehensive review of this drug class, with a primary focus on the three most prominent approved agents: pembrolizumab (Keytruda®), nivolumab (Opdivo®), and cemiplimab (Libtayo®).[4] The clinical success of these antibodies is characterized by their broad efficacy across a diverse array of histologically distinct tumor types, leading to durable responses and unprecedented long-term survival in a subset of patients with metastatic disease. A landmark achievement for this class was the establishment of tissue-agnostic approvals, wherein eligibility for treatment is determined by a predictive genetic biomarker—such as high microsatellite instability (MSI-H) or mismatch repair deficiency (dMMR)—rather than the anatomical site of the tumor's origin.[6]
Concomitant with their unique mechanism of action is a distinct safety profile characterized by a spectrum of inflammatory, autoimmune-like toxicities known as immune-related adverse events (irAEs). These on-target effects can manifest in any organ system and require specialized clinical management, typically involving corticosteroids and interruption of therapy.[4] The field continues to evolve rapidly, with current research focused on elucidating mechanisms of resistance, identifying more precise predictive biomarkers, and developing rational combination therapies that pair anti-PD-1 agents with chemotherapy, radiotherapy, targeted agents, and other immunotherapies. Furthermore, the application of these agents is expanding from the metastatic setting into earlier stages of cancer, including neoadjuvant and adjuvant treatment, with the goal of improving cure rates and preventing disease recurrence.
The PD-1/PD-L1 Immune Checkpoint: A Foundational Mechanism in Oncology
The therapeutic success of anti-PD-1 antibodies is rooted in the elegant manipulation of a fundamental pathway of immune regulation. This pathway, central to maintaining self-tolerance, is frequently co-opted by malignant tumors as a primary mechanism of immune evasion. Understanding its molecular biology is therefore essential to appreciating both the efficacy and the toxicity profile of this drug class.
Molecular Biology of the PD-1 Axis
Programmed Death-1 (PD-1), also known as CD279, is a 288-amino acid transmembrane glycoprotein and a key member of the CD28/CTLA-4 immunoglobulin superfamily.[8] It functions as an inhibitory cell surface receptor, or immune checkpoint, expressed primarily on activated T-cells, B-cells, and natural killer (NK) cells.[9] Its expression is upregulated following T-cell activation, serving as a negative feedback loop to temper the immune response.[11]
The function of PD-1 is mediated through its interaction with two known ligands: Programmed Death-Ligand 1 (PD-L1, also known as B7-H1 or CD274) and Programmed Death-Ligand 2 (PD-L2, also known as B7-DC). PD-L1 is the principal ligand and is broadly expressed on a wide range of hematopoietic and non-hematopoietic cells, including vascular endothelium and pancreatic islets, as well as on many types of tumor cells.[10] Its expression can be either constitutive or inducible, notably upregulated in response to inflammatory cytokines such as interferon-gamma ($IFN-\gamma$), which is often present within the tumor microenvironment (TME).[10] PD-L2 has a more restricted expression pattern, found primarily on professional antigen-presenting cells (APCs) like dendritic cells and macrophages.[1]
Role in Immune Homeostasis
The physiological role of the PD-1/PD-L1 pathway is to function as a critical "off switch" for the immune system. It plays a central part in maintaining peripheral tolerance, which is the process that prevents activated T-cells from attacking the body's own healthy tissues.[1] When an activated T-cell encounters a normal, healthy cell expressing PD-L1 in a peripheral tissue, the binding of PD-L1 to the T-cell's PD-1 receptor delivers a potent inhibitory signal. This signal transduction cascade, involving the recruitment of phosphatases like SHP-2 to the cytoplasmic tail of the PD-1 receptor, effectively dampens T-cell receptor (TCR) signaling. The downstream consequences are the inhibition of T-cell proliferation, a reduction in the production of effector cytokines (such as $IL-2$ and $IFN-\gamma$), and the promotion of T-cell anergy or apoptosis.[3] This mechanism is crucial for resolving inflammation after an infection is cleared and for preventing the development of autoimmune diseases.
Tumor Immune Evasion
Malignant cells have evolved numerous strategies to evade destruction by the host immune system, and the co-option of the PD-1/PD-L1 checkpoint is one of the most significant.[1] Many tumors upregulate the expression of PD-L1 on their cell surface, effectively creating an "immune shield" that mimics the self-tolerance signals of healthy tissue.[14] This can occur through tumor-intrinsic oncogenic signaling pathways or, in a process known as adaptive immune resistance, as a direct response to the local anti-tumor immune response. When tumor-infiltrating lymphocytes (TILs) recognize tumor antigens and release $IFN-\gamma$, this cytokine can induce or further upregulate PD-L1 expression on the cancer cells.[11]
When a cytotoxic T-cell that is primed to attack the tumor engages with a cancer cell, the concurrent binding of the tumor's PD-L1 to the T-cell's PD-1 receptor triggers the inhibitory cascade. This signal overrides the T-cell's activation signals, leading to a state of functional impairment known as T-cell exhaustion. The T-cell becomes anergic, unable to mount an effective cytotoxic response, which allows the tumor to survive and proliferate despite being recognized by the immune system.[1]
Therapeutic Rationale for PD-1 Blockade
The therapeutic strategy of PD-1 blockade is fundamentally restorative. It does not aim to non-specifically stimulate the immune system but rather to unleash a pre-existing, but suppressed, anti-tumor T-cell response.[18] The core concept is to use high-affinity monoclonal antibodies to physically block the PD-1 receptor on T-cells.[3] This blockade prevents PD-L1 and PD-L2 on tumor cells or other cells in the TME from engaging with PD-1. By disrupting this inhibitory interaction, the "brakes" on the T-cell are released.[3] This restores the ability of the previously exhausted or anergic tumor-specific T-cells to execute their effector functions, including proliferation, cytokine secretion, and direct cytotoxic killing of cancer cells.[1]
This mechanism explains why the efficacy of PD-1 blockade is often linked to the baseline immunogenicity of the tumor. Cancers with a high number of mutations (high tumor mutational burden, or TMB) or those with defects in DNA mismatch repair (dMMR/MSI-H) produce a greater number of abnormal proteins, known as neoantigens.[16] These neoantigens are more likely to have been recognized by the immune system, leading to the generation of a pool of tumor-specific T-cells. While these T-cells may have been subsequently suppressed by the PD-1 pathway, they represent a pre-existing army that can be reinvigorated by PD-1 blockade. In essence, the therapy is most effective when there is an underlying, albeit stalled, immune response to restore.
The very same mechanism that confers therapeutic benefit—the disruption of a key self-tolerance checkpoint—is also a double-edged sword that directly predicts the entire class of characteristic side effects. The global blockade of PD-1 can inadvertently allow T-cells to recognize and attack healthy tissues, not just cancer cells. This loss of peripheral tolerance manifests as a unique constellation of inflammatory conditions known as immune-related adverse events (irAEs), which can affect virtually any organ system.[4] These are not off-target toxicities but rather on-target, mechanism-based effects that resemble autoimmune diseases. This unifying principle explains why irAEs such as colitis, pneumonitis, hepatitis, and endocrinopathies are the principal safety concerns for this drug class and why they are managed with immunosuppressive agents like corticosteroids.[7]
Profile of Key Approved Anti-PD-1 Humanized Monoclonal Antibodies
While sharing a common target, the leading approved anti-PD-1 antibodies possess distinct molecular profiles, dosing schedules, and regulatory histories that have shaped their clinical application. The evolution of their development also reflects a maturing understanding of the pharmacology of this drug class, particularly regarding dosing strategies.
Pembrolizumab (Keytruda®)
Pharmacology and Molecular Profile
Pembrolizumab is a high-affinity, humanized monoclonal antibody of the immunoglobulin G4-kappa ($IgG_4-\kappa$) isotype. It is engineered to bind specifically to the human PD-1 receptor and block its interaction with both PD-L1 and PD-L2.[6] The antibody was generated by grafting the variable regions of a murine anti-human PD-1 antibody onto a human $IgG_4$ framework. This framework includes a stabilizing S228P point mutation in the Fc region, which is designed to prevent Fab-arm exchange and minimize antibody-dependent cell-mediated cytotoxicity (ADCC) and other Fc-mediated effector functions.[23]
Administration and Dosing: Pembrolizumab is administered as an intravenous (IV) infusion over 30 minutes.[6] The approved dosing for adults has evolved to offer greater flexibility and patient convenience. The standard regimens are a fixed dose of 200 mg administered every 3 weeks (Q3W) or a fixed dose of 400 mg administered every 6 weeks (Q6W).[26] For pediatric patients, dosing remains weight-based at 2 mg/kg (up to a maximum of 200 mg) every 3 weeks.[25]
Pharmacokinetics (PK): Pembrolizumab is cleared from circulation primarily through non-specific catabolism into small peptides and amino acids, a common pathway for monoclonal antibodies.[28] Its clearance demonstrates a correlation with body weight, which initially supported weight-based dosing. However, subsequent population PK modeling revealed a wide therapeutic index and a flat exposure-response relationship for both efficacy and safety within the clinically tested dose ranges.[29] This crucial finding indicated that once a certain threshold of drug concentration is achieved to ensure saturation of the PD-1 receptors, further increases in exposure do not yield significant gains in efficacy or toxicity. This pharmacological understanding was pivotal in justifying the transition to simpler, more convenient fixed-dose regimens and the approval of the extended-interval 400 mg Q6W schedule, which reduces the frequency of patient visits to infusion centers.[29] Further PK analyses suggest that full target saturation may be achieved at concentrations significantly below the trough levels maintained by current regimens, prompting ongoing discussion about the potential for dose optimization.[30]
Regulatory Landscape and Approved Indications
Pembrolizumab, developed by Merck & Co., received its first approval from the U.S. Food and Drug Administration (FDA) in September 2014 for the treatment of advanced melanoma, marking the first anti-PD-1 therapy approved in the United States.[6] Since then, its clinical development program has been exceptionally broad, leading to approvals for over 30 distinct indications in the U.S. alone.[31]
A landmark moment in oncology occurred in 2017 when the FDA granted pembrolizumab the first-ever "tissue-agnostic" approval. This approval was for the treatment of any unresectable or metastatic solid tumor with the genetic biomarkers of MSI-H or dMMR, regardless of the cancer's anatomical site of origin.[6] This established a new paradigm in cancer therapy, where treatment is guided by a tumor's molecular profile rather than its histology.
The approved indications for pembrolizumab are extensive and continue to expand. Key approvals by major regulatory bodies (FDA, European Medicines Agency [EMA], and Australia's Therapeutic Goods Administration) include, but are not limited to:
- Melanoma: For unresectable or metastatic disease and as adjuvant therapy for resected Stage IIB, IIC, or III melanoma.[32]
- Non-Small Cell Lung Cancer (NSCLC): Across multiple settings, including first-line monotherapy for tumors with high PD-L1 expression, first-line in combination with chemotherapy for all histologies, second-line therapy, and as a perioperative (neoadjuvant followed by adjuvant) treatment for resectable disease.[22]
- Head and Neck Squamous Cell Carcinoma (HNSCC): For metastatic or unresectable, recurrent disease, both as monotherapy and in combination with chemotherapy.[32]
- Classical Hodgkin Lymphoma (cHL): For relapsed or refractory disease in both adult and pediatric patients.[33]
- Urothelial Carcinoma: For locally advanced or metastatic disease in various settings, including for patients ineligible for cisplatin-containing chemotherapy.[6]
- Gastrointestinal Cancers: Including Gastric/Gastroesophageal Junction (GEJ) adenocarcinoma, Esophageal Cancer, and MSI-H/dMMR Colorectal Cancer (CRC).[6]
- Other Malignancies: Including Renal Cell Carcinoma (RCC), Endometrial Carcinoma, Triple-Negative Breast Cancer (TNBC), Cervical Cancer, Hepatocellular Carcinoma (HCC), Malignant Pleural Mesothelioma (MPM), and Merkel Cell Carcinoma (MCC).[6]
Many of these approvals are facilitated by international collaboration, such as the FDA's Project Orbis, which allows for concurrent submission and review among partner agencies like the TGA.[38]
Nivolumab (Opdivo®)
Pharmacology and Molecular Profile
Nivolumab is a fully human monoclonal antibody of the $IgG_4$ isotype that specifically binds to the PD-1 receptor, thereby blocking its interaction with both PD-L1 and PD-L2.[41] As a fully human antibody, it is produced using transgenic mice engineered to have human immunoglobulin genes. Like pembrolizumab, it incorporates a stabilizing S228P mutation in the $IgG_4$ Fc region to prevent Fab-arm exchange and minimize effector functions.[43]
Administration and Dosing: Nivolumab is administered as an IV infusion over 30 to 60 minutes.[44] Its dosing has also transitioned from an initial weight-based regimen (3 mg/kg Q2W) to more convenient flat-dose options for monotherapy, including 240 mg Q2W or 480 mg Q4W.[46] A key feature of nivolumab's clinical use is its frequent application in combination with the anti-CTLA-4 antibody ipilimumab. These combination regimens utilize specific weight-based dosing, such as nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks for four cycles in melanoma, or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four cycles in RCC.[49]
Pharmacokinetics (PK): Nivolumab exhibits linear pharmacokinetics, with dose-proportional increases in Cmax and AUC over the clinically relevant dose range.[51] It has a long elimination half-life of approximately 27 days.[52] Extensive population PK modeling was instrumental in bridging the data from the initial weight-based dosing to the flat-dose and extended-interval regimens. These models demonstrated that a 480 mg Q4W flat dose achieves steady-state trough and average concentrations comparable to the 3 mg/kg Q2W regimen, with no new safety signals identified. This supported the approval of the more convenient Q4W schedule.[48]
Regulatory Landscape and Approved Indications
Nivolumab, developed by Bristol Myers Squibb and Ono Pharmaceutical, was the first PD-1 inhibitor to receive regulatory approval anywhere in the world, with its initial approval in Japan in July 2014 for unresectable melanoma. The U.S. FDA followed with an approval for advanced melanoma in December 2014.[52]
Nivolumab has secured a wide range of indications, many of which are for its use in combination with ipilimumab, establishing dual checkpoint blockade as a standard of care in several cancers. Key approvals from the FDA, EMA, and TGA include:
- Melanoma: For unresectable or metastatic disease (as monotherapy or in combination with ipilimumab) and as adjuvant therapy for resected melanoma.[49]
- Non-Small Cell Lung Cancer (NSCLC): For metastatic disease in the second-line setting, first-line metastatic disease in combination with ipilimumab for tumors with PD-L1 expression $\geq1\%$, and as neoadjuvant treatment with chemotherapy for resectable NSCLC.[49]
- Renal Cell Carcinoma (RCC): As first-line therapy for intermediate/poor-risk patients in combination with ipilimumab, first-line in combination with cabozantinib, and as monotherapy after prior anti-angiogenic therapy.[49]
- Malignant Pleural Mesothelioma (MPM): As first-line therapy in combination with ipilimumab.[49]
- Gastrointestinal Cancers: Including MSI-H/dMMR Colorectal Cancer (in combination with ipilimumab), Hepatocellular Carcinoma (HCC), and various Esophageal, GEJ, and Gastric cancers.[49]
- Other Malignancies: Including Classical Hodgkin Lymphoma (cHL), Squamous Cell Carcinoma of the Head and Neck (SCCHN), and Urothelial Carcinoma.[52]
Cemiplimab (Libtayo®)
Pharmacology and Molecular Profile
Cemiplimab is a recombinant, fully human monoclonal antibody of the $IgG_4$ isotype. It was developed using Regeneron's VelocImmune® technology, which involves mice genetically engineered to produce fully human antibodies.[59] As a fully human antibody, it may theoretically have a lower potential for immunogenicity compared to humanized antibodies. It binds with high affinity to the PD-1 receptor, blocking its interaction with both PD-L1 and PD-L2.[60]
Administration and Dosing: Cemiplimab is administered as a 30-minute IV infusion.[63] The standard recommended dosage is a fixed dose of 350 mg every 3 weeks (Q3W).[63]
Pharmacokinetics (PK): Cemiplimab demonstrates linear and dose-proportional pharmacokinetics across a range of doses. It is cleared via proteolytic catabolism and has a terminal half-life of approximately 22 days.[62] Steady-state concentrations are typically reached after about 4 months of treatment. Population PK modeling has confirmed that the 350 mg Q3W fixed-dose regimen provides consistent and effective drug exposure across various solid tumor types and patient body weights, validating this simple dosing approach.[60]
Regulatory Landscape and Approved Indications
Cemiplimab, co-developed by Regeneron and Sanofi, received its initial FDA approval in September 2018. This approval was a significant milestone as it was the first systemic therapy specifically approved for patients with advanced cutaneous squamous cell carcinoma (CSCC) who are not candidates for curative surgery or radiation.[70]
While its range of indications is not yet as broad as that of pembrolizumab or nivolumab, cemiplimab has established a strong presence in specific clinical niches. Key approvals by the FDA, EMA, and TGA include:
- Cutaneous Squamous Cell Carcinoma (CSCC): For patients with metastatic or locally advanced disease who are not candidates for curative local therapies, and more recently, as adjuvant treatment for patients at high risk of recurrence after surgery and radiation.[59]
- Basal Cell Carcinoma (BCC): For patients with locally advanced or metastatic BCC who have progressed on or are intolerant to a hedgehog pathway inhibitor.[66]
- Non-Small Cell Lung Cancer (NSCLC): As a first-line monotherapy for patients with advanced NSCLC whose tumors have high PD-L1 expression (TPS $\geq50\%$) and no targetable genomic aberrations, and as a first-line treatment in combination with platinum-based chemotherapy for patients regardless of PD-L1 status.[66]
- Cervical Cancer: For patients with recurrent or metastatic cervical cancer whose disease has progressed on or after chemotherapy (EMA approval).[72]
Table 1: Comparative Overview of Major Anti-PD-1 Humanized Monoclonal Antibodies
| Feature | Pembrolizumab (Keytruda®) | Nivolumab (Opdivo®) | Cemiplimab (Libtayo®) |
|---|
| Molecular Type | Humanized $IgG_4-\kappa$ | Fully Human $IgG_4$ | Fully Human $IgG_4$ |
| Mechanism of Action | Binds to PD-1, blocking interaction with PD-L1 and PD-L2 6 | Binds to PD-1, blocking interaction with PD-L1 and PD-L2 41 | Binds to PD-1, blocking interaction with PD-L1 and PD-L2 60 |
| Standard Dosing (Monotherapy) | 200 mg IV Q3W or 400 mg IV Q6W 26 | 240 mg IV Q2W or 480 mg IV Q4W 47 | 350 mg IV Q3W 63 |
| Key Landmark Approval | First tissue-agnostic approval (for MSI-H/dMMR solid tumors) 6 | First PD-1 inhibitor approved globally (in Japan, 2014) 44 | First systemic therapy approved for advanced CSCC 70 |
| Notable Combination Regimens | With chemotherapy (e.g., NSCLC, HNSCC); with axitinib (RCC); with lenvatinib (Endometrial) 6 | With ipilimumab (anti-CTLA-4) for Melanoma, RCC, NSCLC, MPM, CRC, HCC, OSCC 44 | With platinum-based chemotherapy for NSCLC 66 |
Clinical Efficacy Across Major Tumor Types
The clinical development of anti-PD-1 antibodies has yielded transformative results across a wide spectrum of malignancies. The data from pivotal, large-scale clinical trials not only established the superiority of these agents over previous standards of care but also introduced the concept of long-term, durable survival for a meaningful subset of patients with metastatic disease. This section reviews the landmark efficacy data in several key indications.
Metastatic Melanoma
Melanoma served as the foundational tumor type for demonstrating the profound efficacy of immune checkpoint blockade.
- Pembrolizumab: The KEYNOTE-006 trial was a pivotal phase 3 study that directly compared pembrolizumab with the anti-CTLA-4 antibody ipilimumab, the then-standard immunotherapy for first-line treatment. The results were definitive, showing that pembrolizumab provided superior overall survival (OS) and progression-free survival (PFS).[74] A long-term follow-up analysis at 5 years reported a median OS of 32.7 months for patients receiving pembrolizumab, compared to just 15.9 months for those receiving ipilimumab.[74] Furthermore, the objective response rate (ORR)—the proportion of patients with significant tumor shrinkage—was substantially higher with pembrolizumab at 33% (including a 6% complete response rate) versus 12% for ipilimumab.[75] These data firmly established anti-PD-1 monotherapy as a superior first-line standard of care.
- Nivolumab: The CheckMate-067 trial provided a landmark three-arm comparison of nivolumab monotherapy, ipilimumab monotherapy, and the combination of nivolumab plus ipilimumab in previously untreated advanced melanoma patients. The results, followed for an unprecedented 10 years, have reshaped expectations for long-term survival in this disease.[76] The final analysis revealed a striking divergence in outcomes: the median OS was nearly six years for the combination therapy group, a remarkable achievement. This compared favorably to three years for nivolumab monotherapy and only 20 months for ipilimumab monotherapy. Most profoundly, the 10-year melanoma-specific survival rate for patients who initiated treatment with the nivolumab and ipilimumab combination exceeded 50%, a figure that allows clinicians to begin discussing the potential for a functional cure in a disease that was almost uniformly fatal just over a decade ago.[76]
Non-Small Cell Lung Cancer (NSCLC)
The success of anti-PD-1 therapy in NSCLC, the leading cause of cancer death globally, has been particularly impactful, demonstrating benefit in both squamous and non-squamous histologies and across various lines of therapy.
- Pembrolizumab: The phase 1b KEYNOTE-001 trial was the first study to demonstrate the long-term potential of PD-1 blockade in NSCLC. Updated results with a median follow-up of over five years showed a 5-year OS rate of 23.2% in treatment-naïve patients and 15.5% in previously treated patients.[77] These figures represented a dramatic improvement over the historical 5-year survival rate of approximately 5% for metastatic NSCLC.[77] The 10-year follow-up data continued to confirm this durable survival benefit for a subset of patients, underscoring the therapy's long-lasting impact.[79] This early success paved the way for phase 3 trials that established pembrolizumab, either as monotherapy for tumors with high PD-L1 expression or in combination with chemotherapy, as the global standard of care for first-line metastatic NSCLC.
- Nivolumab: The CheckMate 017 (squamous histology) and CheckMate 057 (non-squamous histology) trials were pivotal phase 3 studies that established nivolumab's superiority over the standard second-line chemotherapy, docetaxel, in patients with previously treated advanced NSCLC. A pooled analysis with a minimum 5-year follow-up demonstrated a profound and sustained survival benefit. The 5-year OS rate for patients treated with nivolumab was 13.4%, a more than five-fold increase compared to the 2.6% rate observed in the docetaxel arm.[80] The 2-year OS rates were 23% for nivolumab versus 8% for docetaxel in squamous NSCLC, and 29% versus 16% in non-squamous NSCLC, respectively.[82] A critical observation from this long-term data was the durability of response; patients who were progression-free at the 3-year mark had a 93% probability of being alive at 5 years, highlighting the stability of the benefit in long-term responders.[80]
- Cemiplimab: Cemiplimab has also demonstrated significant efficacy in NSCLC. The EMPOWER-Lung 1 trial established its role as a first-line monotherapy, showing superior OS and PFS compared to chemotherapy in patients whose tumors had high PD-L1 expression (TPS $\geq50\%$).[84] For a broader patient population, the EMPOWER-Lung 3 trial evaluated cemiplimab in combination with chemotherapy versus chemotherapy alone as a first-line treatment. The 5-year results were impressive, showing that the combination more than doubled the 5-year survival rate to 19.4% versus 8.8% for chemotherapy alone. The median OS was 21.1 months for the combination versus 12.9 months for chemotherapy.[85]
Cutaneous Squamous Cell Carcinoma (CSCC)
- Cemiplimab: Advanced CSCC, particularly in patients not amenable to curative surgery or radiation, was a disease with limited systemic treatment options and a poor prognosis. The pivotal, single-arm, phase 2 EMPOWER-CSCC 1 trial established cemiplimab as a highly effective new standard of care. Long-term follow-up data demonstrated an ORR of 46-47%, with a notable 16% of patients achieving a complete response.[86] The most striking feature of the results was the durability of these responses. The median duration of response (DOR) and the median OS were not reached even after several years of follow-up. The estimated probability of OS at 24 months was 73.3%, a testament to the profound and lasting benefit cemiplimab provides in this patient population.[87]
The clinical data across these diverse tumor types reveals a clear strategic evolution in the application of anti-PD-1 therapy. These agents were first validated as a salvage option in heavily pretreated patients who had exhausted standard therapies.[77] The remarkable success in that setting propelled their investigation in first-line trials, where they consistently demonstrated superiority over chemotherapy, establishing a new standard of care.[32] The next logical frontier, now well underway, is the movement of these highly effective therapies into earlier stages of disease. Adjuvant trials, designed to prevent recurrence after surgery, and neoadjuvant trials, which use immunotherapy before surgery to shrink tumors and prime a systemic anti-tumor immune response, are demonstrating significant benefits.[22] This trajectory reflects a growing confidence in the efficacy and manageable safety of PD-1 blockade, with the ultimate goal of increasing cure rates by applying the most effective treatments earlier in the disease course.
A second critical theme emerging from the long-term follow-up data is the phenomenon of the "tail on the curve." In historical survival plots for metastatic solid tumors treated with chemotherapy, the curves typically decline steadily toward zero. In stark contrast, the 5- and 10-year survival curves from trials like KEYNOTE-001 and CheckMate-067 exhibit a distinct plateau, or "tail," after the first few years.[76] This indicates that a subset of patients who achieve a durable response to immunotherapy enter a state of long-term disease control, with a significantly reduced risk of subsequent relapse or death from their cancer. This has profound implications, allowing clinicians and patients to discuss the possibility of long-term remission and even functional cure—a conversation that was largely impossible for most metastatic solid tumors in the pre-immunotherapy era.[76]
Table 2: Summary of Efficacy Data from Pivotal Trials by Indication
| Indication | Trial Name | Drug(s) | Patient Population | Control Arm | Median OS (Drug vs. Control) | Hazard Ratio (OS) | 5-Year OS Rate (%) | ORR (%) |
|---|
| Melanoma | KEYNOTE-006 | Pembrolizumab | 1L, Advanced | Ipilimumab | 32.7 mo vs. 15.9 mo | 0.73 | N/A (at 5 yrs) | 33% vs. 12% |
| Melanoma | CheckMate-067 | Nivo + Ipi | 1L, Advanced | Ipilimumab | 72.1 mo vs. 19.9 mo | 0.52 | 52% vs. 26% (at 7.5 yrs) | 58% vs. 19% |
| Melanoma | CheckMate-067 | Nivolumab | 1L, Advanced | Ipilimumab | 36.9 mo vs. 19.9 mo | 0.65 | 44% vs. 26% (at 7.5 yrs) | 44% vs. 19% |
| NSCLC (2L+) | KEYNOTE-001 | Pembrolizumab | Previously Treated | Single Arm | 10.5 mo | N/A | 15.5% | 18% |
| NSCLC (1L) | KEYNOTE-001 | Pembrolizumab | Treatment-Naïve | Single Arm | 22.3 mo | N/A | 23.2% | 24.8% |
| NSCLC (2L+) | CheckMate 017/057 | Nivolumab | Previously Treated | Docetaxel | 11.1 mo vs. 8.1 mo | 0.68 | 13.4% vs. 2.6% | N/A |
| NSCLC (1L) | EMPOWER-Lung 3 | Cemiplimab + Chemo | 1L, Advanced | Chemotherapy | 21.1 mo vs. 12.9 mo | 0.66 | 19.4% vs. 8.8% | 43.6% vs. 22.1% |
| CSCC | EMPOWER-CSCC 1 | Cemiplimab | Advanced | Single Arm | Not Reached | N/A | 73.3% (at 2 yrs) | 46.1% |
| 74 | | | | | | | |
Predictive Biomarkers for Patient Stratification
Despite the broad efficacy of anti-PD-1 therapies, a substantial portion of patients do not respond. This has driven an intensive search for predictive biomarkers to identify which patients are most likely to benefit, thereby optimizing treatment selection, avoiding unnecessary toxicity, and managing healthcare costs. While no single biomarker is perfect, several have been incorporated into clinical practice.
PD-L1 Expression
The expression level of PD-L1 on tumor cells or associated immune cells is the most widely implemented biomarker for anti-PD-1 therapy. The underlying rationale is straightforward: tumors that have upregulated PD-L1 are likely more dependent on the PD-1/PD-L1 axis for immune evasion, and therefore, blocking this interaction should be more impactful.[90]
However, the clinical application of PD-L1 testing is fraught with complexity. Several different immunohistochemistry (IHC) assays have been developed, each using a different antibody clone (e.g., 22C3 for pembrolizumab, 28-8 for nivolumab) and often paired with a specific drug. Furthermore, scoring methodologies vary. The Tumor Proportion Score (TPS) measures the percentage of viable tumor cells showing partial or complete membrane staining, while the Combined Positive Score (CPS) is a ratio of all PD-L1 staining cells (tumor cells, lymphocytes, macrophages) to the total number of viable tumor cells, multiplied by 100.[32] Different tumor types and clinical settings use different scoring systems and positivity cutoffs (e.g., TPS $\geq1\%$, TPS $\geq50\%$, or CPS $\geq1$, CPS $\geq10$) to define eligibility for treatment.[32]
Critically, PD-L1 expression is an imperfect and dynamic biomarker. While numerous clinical trials have shown a positive correlation between higher PD-L1 expression and improved response rates and survival, the predictive value is not absolute.[91] A meaningful number of patients with PD-L1-negative tumors still derive clinical benefit, and conversely, many patients with PD-L1-positive tumors do not respond.[90] This inconsistency stems from several factors, including intra-tumoral and inter-tumoral heterogeneity of PD-L1 expression, the dynamic nature of its expression in response to the immune microenvironment, and the technical variability of the assays themselves.[11] Thus, while PD-L1 is a useful enrichment tool, it is not a definitive binary predictor.
Tumor Mutational Burden (TMB)
Tumor Mutational Burden (TMB) is a quantitative measure of the total number of somatic mutations per megabase of DNA in a tumor's genome. The central hypothesis is that a higher TMB leads to the generation of a greater number of mutant proteins, which can be processed and presented as "neoantigens" on the tumor cell surface. A higher neoantigen load increases the probability that the tumor will be recognized as foreign by the immune system, rendering it more immunogenic and thus more susceptible to the effects of immune checkpoint blockade.[16]
TMB has emerged as a powerful biomarker, often independent of PD-L1 status, particularly in cancers like NSCLC and melanoma.[16] In 2020, the FDA granted an accelerated, tissue-agnostic approval to pembrolizumab for the treatment of patients with unresectable or metastatic TMB-high (TMB-H, defined as $\geq10$ mutations/megabase) solid tumors that have progressed following prior treatment.[35] However, the utility of TMB is not universal. Comprehensive analyses have shown that while TMB is strongly predictive in some cancers, it has little to no predictive value in others, such as breast, prostate, and brain cancers.[96] This suggests that in certain tumor types, other immune resistance mechanisms may be dominant and can override the potential benefit of a high neoantigen load.
Mismatch Repair Deficiency (dMMR)/Microsatellite Instability-High (MSI-H)
Tumors with a deficient DNA mismatch repair (dMMR) system are unable to correct errors that occur during DNA replication. This leads to the accumulation of thousands of mutations, particularly in repetitive DNA sequences known as microsatellites, a state referred to as microsatellite instability-high (MSI-H). Consequently, dMMR/MSI-H tumors have an exceptionally high TMB and a rich neoantigen landscape.
This genetic signature makes these tumors highly immunogenic and exquisitely sensitive to PD-1 blockade. dMMR/MSI-H status is one of the most robust predictive biomarkers for response to immunotherapy. Its strong predictive power across numerous different cancer types led to the landmark 2017 FDA approval of pembrolizumab for any dMMR/MSI-H solid tumor, the first approval based solely on a biomarker irrespective of cancer origin.[6]
The current biomarker landscape reveals that "tumor immunogenicity" is not a monolithic entity but a complex and multi-faceted property. Each of the primary biomarkers—PD-L1, TMB, and MSI-H—captures a different aspect of the intricate dance between the tumor and the immune system. PD-L1 expression reflects an active, adaptive immune resistance mechanism within the TME. TMB quantifies the underlying potential for neoantigen formation, a prerequisite for immune recognition. MSI-H represents an extreme phenotype of TMB driven by a specific genetic defect. The fact that their predictive utility varies by cancer type indicates that the dominant mechanisms of immune evasion are context-dependent. For instance, in a "cold" or non-inflamed tumor, a high TMB may be insufficient to generate a response if there are no T-cells present to recognize the neoantigens. This understanding is driving the field away from a search for a single "best" biomarker and toward multi-modal approaches that integrate genomics (TMB, MSI-H), protein expression (PD-L1), and potentially transcriptomics (gene expression signatures) to create a more holistic and accurate predictive model of the tumor-immune ecosystem for each individual patient.[95]
Safety Profile and Management of Immune-Related Adverse Events (irAEs)
The unique mechanism of action of anti-PD-1 antibodies gives rise to a distinct and characteristic profile of side effects. These toxicities, termed immune-related adverse events (irAEs), are fundamentally different from the cytotoxic side effects of traditional chemotherapy and require a specialized approach to recognition and management.
Spectrum of irAEs
Immune-related adverse events are a direct, on-target consequence of augmenting the immune system. By blocking a key checkpoint that maintains self-tolerance, these therapies can lead to the activation of T-cells that recognize self-antigens, resulting in inflammatory, autoimmune-like reactions in healthy organs and tissues.[4] IrAEs can affect any organ system and can occur at any point during treatment, and in some cases, even months after the cessation of therapy.[20]
Common irAEs: The most frequently observed irAEs are generally mild to moderate in severity. These include:
- Dermatologic: Rash (maculopapular, pruritic) and pruritus (itching) are among the most common, occurring in 15-25% of patients.[62]
- Gastrointestinal: Diarrhea is common, reported in approximately 20-27% of patients. In more severe forms, this can progress to colitis.[67]
- General: Fatigue is the most common side effect of any kind, reported in roughly 30% of patients receiving monotherapy.[101]
- Endocrine: Endocrinopathies are a hallmark toxicity. Hypothyroidism is the most frequent, occurring in up to 9% of patients, followed by hyperthyroidism.[62] Hypophysitis and adrenal insufficiency are less common but can be serious.[62]
- Musculoskeletal: Arthralgia and myalgia are also frequently reported.[42]
Serious but Less Common irAEs: While less frequent, severe (Grade 3-4) or fatal irAEs can occur and require immediate medical attention. These include:
- Pneumonitis: Inflammation of the lungs, presenting with cough, dyspnea, and chest pain. It occurs in approximately 3-9% of patients, with severe cases in 1-3%.[67] The risk is higher in patients with prior thoracic radiation.[104]
- Hepatitis: Immune-mediated liver inflammation, detected by elevated transaminases (AST/ALT). Severe hepatitis occurs in 1-2% of patients.[103]
- Myocarditis: Inflammation of the heart muscle. Although rare (<1%), it can be fulminant and fatal, presenting with chest pain, arrhythmias, or heart failure symptoms.[103]
- Nephritis: Inflammation of the kidneys, leading to renal dysfunction, typically presenting as a rising creatinine level.[42]
- Neurologic: A range of rare but serious toxicities including encephalitis, Guillain-Barré syndrome, and myasthenia gravis.[42]
- Severe Skin Reactions: Including Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), which are medical emergencies.[62]
Grading and Management Principles
The management of irAEs is guided by their severity, which is graded according to the National Cancer Institute's Common Terminology Criteria for Adverse Events (CTCAE). The fundamental principles of management, as outlined in guidelines from organizations like the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO), are early recognition, prompt intervention, and modulation of the immune response, primarily with corticosteroids.[7]
A general, grade-based management algorithm is as follows:
- Grade 1 (Mild): For most Grade 1 toxicities, anti-PD-1 therapy can be continued with close monitoring and symptomatic treatment (e.g., topical corticosteroids for rash, antidiarrheals for mild diarrhea).[7]
- Grade 2 (Moderate): Treatment should be held. For persistent or symptomatic toxicities, systemic corticosteroids (e.g., prednisone at a dose of 0.5 to 1 mg/kg/day or equivalent) should be initiated. Anti-PD-1 therapy can be considered for resumption once symptoms and/or laboratory values have improved to Grade 1 or less and the patient is on a low dose of corticosteroids.[7]
- Grade 3 (Severe): Treatment should be held, and high-dose systemic corticosteroids (e.g., prednisone at 1 to 2 mg/kg/day or IV methylprednisolone) should be started immediately. Patients often require hospitalization. Once symptoms improve, the corticosteroids must be tapered very slowly, typically over a period of at least 4 to 6 weeks, to prevent recurrence. For certain irAEs that are refractory to steroids (e.g., colitis), additional immunosuppressive agents like infliximab may be required. Permanent discontinuation of the anti-PD-1 agent is often necessary.[7]
- Grade 4 (Life-threatening): Anti-PD-1 therapy must be permanently discontinued. Patients require immediate hospitalization and treatment with high-dose IV corticosteroids. The main exception is for endocrinopathies (e.g., hypothyroidism, type 1 diabetes) that can be controlled with permanent hormone replacement therapy.[7]
Given the potential for rapid escalation of these toxicities, patient and caregiver education is a cornerstone of safe treatment. Patients must be thoroughly counseled before starting therapy about the signs and symptoms of common and serious irAEs and instructed to report any new or worsening symptoms to their clinical team immediately.[21]
Table 3: Guideline for Management of Common Immune-Related Adverse Events (irAEs)
| Organ System / irAE | Key Symptoms | Diagnostic Workup | Management by Grade |
|---|
| Pneumonitis | New or worsening cough, shortness of breath, chest pain, fever | Chest imaging (X-ray or high-resolution CT), rule out infection, bronchoscopy if needed | Grade 2: Hold therapy; start prednisone 1-2 mg/kg/day.Grade 3/4: Permanently discontinue; hospitalize; start IV methylprednisolone 1-2 mg/kg/day. Taper steroids over $\geq4-6$ weeks.21 |
| Colitis / Diarrhea | Diarrhea, abdominal pain, blood or mucus in stool, fever | Stool studies to rule out infection (especially C. difficile, CMV), endoscopy with biopsy for severe/refractory cases | Grade 2: Hold therapy; start prednisone 0.5-1 mg/kg/day if no improvement with symptomatic care.Grade 3/4: Permanently discontinue; hospitalize; start IV methylprednisolone 1-2 mg/kg/day. If no improvement, consider infliximab.21 |
| Hepatitis | Often asymptomatic; fatigue, nausea, jaundice (late sign) | Monitor AST, ALT, bilirubin at baseline and before each dose. Rule out viral hepatitis, other causes. | Grade 2: Hold therapy; start prednisone 0.5-1 mg/kg/day.Grade 3/4: Permanently discontinue; start prednisone 1-2 mg/kg/day. For refractory cases, consider mycophenolate mofetil.27 |
| Endocrinopathies | Hypo/Hyperthyroidism: Fatigue, weight changes, temperature intolerance. Hypophysitis: Headache, visual changes, fatigue, hypotension. Adrenal Insufficiency: Severe fatigue, nausea, hypotension. | Thyroid function tests (TSH, free T4), cortisol, ACTH, pituitary hormones, MRI of pituitary for suspected hypophysitis | Hypo/Hyperthyroidism: Manage with hormone replacement (levothyroxine) or anti-thyroid drugs. Can often continue immunotherapy.Symptomatic Grade 2+ Hypophysitis/Adrenal Insufficiency: Hold therapy; start prednisone 1-2 mg/kg/day and appropriate hormone replacement. Taper steroids once stable.21 |
| Dermatitis / Rash | Rash (maculopapular), severe pruritus (itch) | Physical exam, skin biopsy for severe or atypical cases to rule out SJS/TEN | Grade 1/2: Continue or hold therapy; manage with topical corticosteroids, oral antihistamines.Grade 3: Hold therapy; start oral prednisone 0.5-1 mg/kg/day.Grade 4 (SJS/TEN): Permanently discontinue; hospitalize for urgent dermatology consult and supportive care.21 |
| (Management principles synthesized from ASCO/ESMO guidelines and prescribing information 7) | | | |
Future Perspectives and Strategic Recommendations
The field of anti-PD-1 therapy is in a state of continuous and rapid evolution. While monotherapy has established a new foundation for cancer treatment, the focus has now shifted toward overcoming resistance, refining patient selection, and intelligently combining therapies to further improve outcomes.
Overcoming Resistance to PD-1 Blockade
A primary clinical challenge is that a significant proportion of patients, between 30% and 60%, exhibit primary (innate) resistance and do not respond to anti-PD-1 monotherapy. Additionally, some initial responders may later develop secondary (acquired) resistance.[12] Research has identified several key mechanisms contributing to this resistance:
- T-cell Exclusion: Some tumors are "cold" or non-inflamed, lacking a pre-existing T-cell infiltrate. In these cases, there are no effector T-cells within the TME for PD-1 blockade to reinvigorate.[12]
- T-cell Exhaustion: In some tumors, T-cells are so profoundly exhausted that simply blocking PD-1 is insufficient to restore their function. They may co-express other inhibitory checkpoint receptors, such as CTLA-4, LAG-3, and TIM-3.[9]
- Immunosuppressive TME: The tumor microenvironment can be populated by other immunosuppressive cell types, such as myeloid-derived suppressor cells (MDSCs) and regulatory T-cells (Tregs), which use mechanisms other than the PD-1 pathway to inhibit anti-tumor immunity.[12]
- Tumor-Intrinsic Factors: Resistance can also be driven by factors within the cancer cell itself, such as mutations in the $IFN-\gamma$ signaling pathway, which prevent the tumor from being recognized by T-cells, or activation of oncogenic pathways like WNT/$\beta$-catenin that contribute to immune exclusion.[12]
Novel Combination Strategies
The leading strategy to overcome these resistance mechanisms and enhance the efficacy of PD-1 blockade is through rational combination therapy. The goal is to target multiple, non-redundant pathways of immune suppression or to make "cold" tumors "hot."
- Dual Checkpoint Blockade: The combination of an anti-PD-1 antibody (working within the TME) and an anti-CTLA-4 antibody (working primarily in lymph nodes to prime T-cells) has demonstrated synergistic activity. The nivolumab plus ipilimumab combination has shown superior efficacy over monotherapy in melanoma, RCC, and NSCLC, among others, establishing this as a powerful, albeit more toxic, treatment option.[107] The approval of the nivolumab plus relatlimab (an anti-LAG-3 antibody) combination for melanoma represents the next wave of dual checkpoint blockade targeting different exhaustion pathways.[4]
- Chemo-immunotherapy: Combining PD-1 inhibitors with conventional chemotherapy has become a first-line standard of care in many cancers, including NSCLC. The rationale is multi-faceted: chemotherapy provides rapid tumor debulking, and by inducing immunogenic cell death, it can increase the release and presentation of tumor antigens, thereby enhancing the T-cell response that PD-1 blockade can then unleash.[22]
- Combinations with Radiotherapy (RT): RT is being actively investigated in combination with PD-1 blockade. Localized radiation can act as an in situ vaccine, promoting antigen release and recruiting T-cells to the irradiated tumor. This can potentially convert a "cold" tumor into a "hot" one and, in some cases, lead to an "abscopal effect," where the localized treatment induces a systemic immune response that attacks distant, non-irradiated metastases.[12]
Emerging Research and Development
The future of immuno-oncology will be shaped by innovations in drug delivery, novel therapeutic modalities, and more sophisticated biomarker strategies.
- New Formulations: To improve patient convenience and reduce the burden on healthcare systems, subcutaneous (SC) formulations of both pembrolizumab and nivolumab are in late-stage development. These formulations, which can be administered in minutes rather than via a 30-60 minute IV infusion, are expected to significantly improve the treatment experience.[110]
- Small Molecule Inhibitors: While monoclonal antibodies have been transformative, research is progressing on orally bioavailable small molecule inhibitors that can disrupt the PD-1/PD-L1 protein-protein interaction. These agents could offer the major advantages of oral administration, lower manufacturing costs, and potentially different pharmacokinetic and tissue penetration profiles compared to large-molecule antibodies.[112]
- Advanced Biomarkers and Personalized Therapy: The limitations of current biomarkers are driving research toward more comprehensive and dynamic predictors of response. The future likely involves multi-modal approaches that integrate genomics (TMB, MSI-H), transcriptomics (gene expression profiling to characterize the TME), proteomics, and even analysis of the gut microbiome, which has been shown to influence immunotherapy outcomes.[94]
This trajectory indicates that the field is moving away from a "one-size-fits-all" approach. The future of immuno-oncology lies in a highly personalized, multi-modal strategy. The goal is to use advanced diagnostics to understand the specific immune resistance mechanisms at play in an individual patient's tumor—be it a lack of T-cell infiltration, the presence of specific inhibitory cells, or a defect in antigen presentation. Based on this "immune diagnosis," clinicians will be able to select the most rational and intelligent sequence or combination of therapies—chemotherapy, RT, targeted agents, and multiple immunotherapies—to precisely counteract those resistance mechanisms and maximize the probability of a durable clinical response. This represents the true paradigm of personalized immuno-oncology.
Conclusion
Recombinant anti-PD-1 humanized monoclonal antibodies have irrevocably transformed the field of oncology. By targeting a fundamental mechanism of tumor immune evasion, agents such as pembrolizumab, nivolumab, and cemiplimab have delivered unprecedented and durable survival benefits to patients across a broad and growing range of malignancies. The long-term follow-up from pivotal clinical trials, demonstrating a plateau in survival curves, has introduced the concept of functional cure for a subset of patients with previously fatal metastatic diseases.
The successful clinical application of these therapies has been paralleled by significant scientific progress. The evolution from weight-based to fixed-dose and extended-interval regimens reflects a sophisticated understanding of their pharmacokinetics, enhancing both patient convenience and healthcare efficiency. The validation of predictive biomarkers like PD-L1, TMB, and particularly MSI-H, has ushered in an era of precision immuno-oncology, culminating in landmark tissue-agnostic approvals that prioritize molecular biology over traditional histology.
However, significant challenges remain. The management of immune-related adverse events, a direct consequence of the therapy's mechanism of action, requires constant vigilance and specialized expertise. More critically, overcoming both primary and acquired resistance to PD-1 blockade is the foremost challenge. The path forward lies in the continued pursuit of rational combination therapies that target multiple, non-redundant immunosuppressive pathways. The integration of anti-PD-1 agents with chemotherapy, radiotherapy, other checkpoint inhibitors, and novel targeted agents, guided by increasingly sophisticated multi-modal biomarkers, will be key to extending the benefits of immunotherapy to a larger proportion of patients. The ongoing development of novel formulations and therapeutic modalities promises to further refine and expand the utility of this revolutionary drug class, solidifying its role as a cornerstone of modern cancer care.
Works cited
- PD-1 and PD-L1: architects of immune symphony and immunotherapy breakthroughs in cancer treatment - Frontiers, accessed October 23, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1296341/full
- PD-1 and PD-L1 inhibitors - Wikipedia, accessed October 23, 2025, https://en.wikipedia.org/wiki/PD-1_and_PD-L1_inhibitors
- Immune Checkpoint Inhibitors - NCI, accessed October 23, 2025, https://www.cancer.gov/about-cancer/treatment/types/immunotherapy/checkpoint-inhibitors
- Immune Checkpoint Inhibitors and Their Side Effects | American Cancer Society, accessed October 23, 2025, https://www.cancer.org/cancer/managing-cancer/treatment-types/immunotherapy/immune-checkpoint-inhibitors.html
- Key Developments in PD-(L)1 Inhibitors - DelveInsight, accessed October 23, 2025, https://www.delveinsight.com/blog/key-developments-in-pd-l1-inhibitors
- Pembrolizumab - Wikipedia, accessed October 23, 2025, https://en.wikipedia.org/wiki/Pembrolizumab
- Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/34724392/
- The PD-1/PD-L1 Pathway: A Perspective on Comparative Immuno-Oncology - MDPI, accessed October 23, 2025, https://www.mdpi.com/2076-2615/12/19/2661
- PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations - PMC - PubMed Central, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6605868/
- The Evolving Landscape of Biomarkers for Anti-PD-1 or Anti-PD-L1 Therapy - MDPI, accessed October 23, 2025, https://www.mdpi.com/2077-0383/8/10/1534
- PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy - AACR Journals, accessed October 23, 2025, https://aacrjournals.org/mct/article/14/4/847/91937/PD-L1-Expression-as-a-Predictive-Biomarker-in
- Future of anti-PD-1/PD-L1 applications: Combinations with other therapeutic regimens, accessed October 23, 2025, https://www.cjcrcn.org/article/html_9792.html
- pmc.ncbi.nlm.nih.gov, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7136921/#:~:text=PD%2D1%2FPD%2DL1%20pathway%20controls%20the%20induction%20and,immune%20responses%20(Figure%201).
- How Keytruda Cancer Treatment Works (Update) - YouTube, accessed October 23, 2025, https://www.youtube.com/watch?v=R0ccmAXq4RU
- Cemiplimab - AIM with Immunotherapy, accessed October 23, 2025, https://aimwithimmunotherapy.org/wp-content/uploads/2025/01/IO-Cemiplimab-HCPToolkit-2024.pdf
- Tumor mutational burden (TMB) as a biomarker of response to immunotherapy in small cell lung cancer - Journal of Thoracic Disease - AME Publishing Company, accessed October 23, 2025, https://jtd.amegroups.org/article/view/23082/html
- Immunological Mechanisms behind Anti-PD-1/PD-L1 Immune Checkpoint Blockade: Intratumoral Reinvigoration or Systemic Induction? - MDPI, accessed October 23, 2025, https://www.mdpi.com/2227-9059/12/4/764
- Anti–PD-1/PD-L1 therapy of human cancer: past, present, and future - PMC - NIH, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4588282/
- The Challenges of Tumor Mutational Burden as an Immunotherapy, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7878292/
- 1 Treatment Guideline: Basic Principles for Management of Immune-related Adverse Events (IrAEs) Caused by immunotherapy, accessed October 23, 2025, https://www.gloshospitals.nhs.uk/documents/6963/Immunotherapy_Treatment_Guidelines-Basic_Principles_for_management_of_IRAEs.pdf
- Management of Immune-Related Adverse Events in Patients ..., accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6481621/
- FDA Approves KEYTRUDA® (pembrolizumab) for Treatment of Patients With Resectable (T≥4 cm or N+) NSCLC in Combination With Chemotherapy as Neoadjuvant Treatment, Then Continued as a Single Agent as Adjuvant Treatment After Surgery - Merck.com, accessed October 23, 2025, https://www.merck.com/news/fda-approves-keytruda-pembrolizumab-for-treatment-of-patients-with-resectable-t%E2%89%A54-cm-or-n-nsclc-in-combination-with-chemotherapy-as-neoadjuvant-treatment-then-continued-as-a-single/
- Pembrolizumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 23, 2025, https://go.drugbank.com/drugs/DB09037
- Immunotherapy for Metastatic Melanoma - Pembrolizumab, Keytruda - MRA, accessed October 23, 2025, https://www.curemelanoma.org/patient-eng/melanoma-treatment/options/keytruda-pembrolizumab
- Keytruda Dosage: Form, Strength, How to Use, and More - Healthline, accessed October 23, 2025, https://www.healthline.com/health/drugs/keytruda-dosage
- Dosing Schedule for KEYTRUDA® (pembrolizumab) | HCP, accessed October 23, 2025, https://www.keytrudahcp.com/dosing/options/
- Keytruda (pembrolizumab) dosing, indications, interactions, adverse effects, and more, accessed October 23, 2025, https://reference.medscape.com/drug/keytruda-pembrolizumab-999962
- Evaluation of the pharmacokinetics and metabolism of pembrolizumab in the treatment of melanoma - PMC - PubMed Central, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5613934/
- Utilization of pharmacokinetics in the dosing of pembrolizumab in non-small cell lung cancer, accessed October 23, 2025, https://actr.amegroups.org/article/view/8868/html
- Case report: Pharmacokinetics of pembrolizumab in a ... - Frontiers, accessed October 23, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.960116/full
- FDA Approves KEYTRUDA® (pembrolizumab) for Treatment of Patients With High-Risk Early-Stage Triple-Negative Breast Cancer in Combination With Chemotherapy as Neoadjuvant Treatment, Then Continued as Single Agent as Adjuvant Treatment After Surgery - Merck.com, accessed October 23, 2025, https://www.merck.com/news/fda-approves-keytruda-pembrolizumab-for-treatment-of-patients-with-high-risk-early-stage-triple-negative-breast-cancer-in-combination-with-chemotherapy-as-neoadjuvant-treatment-then-continued/
- Approved Indications for KEYTRUDA® (pembrolizumab), accessed October 23, 2025, https://www.keytrudahcp.com/approved-indications/
- Keytruda | European Medicines Agency (EMA), accessed October 23, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/keytruda
- This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed October 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125514s040lbl.pdf
- Keytruda - SEER*Rx Interactive Antineoplastic Drugs Database, accessed October 23, 2025, https://seer.cancer.gov/seertools/seerrx/rx/5453af97e4b03ba322415853/
- KEYTRUDA (Merck Sharp & Dohme (Australia) Pty Ltd), accessed October 23, 2025, https://www.tga.gov.au/resources/prescription-medicines-registrations/keytruda-merck-sharp-dohme-australia-pty-ltd-25
- keytruda-epar-product-information_en.pdf - European Medicines Agency, accessed October 23, 2025, https://www.ema.europa.eu/en/documents/product-information/keytruda-epar-product-information_en.pdf
- FDA approves pembrolizumab with chemotherapy for unresectable advanced or metastatic malignant pleural mesothelioma, accessed October 23, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-chemotherapy-unresectable-advanced-or-metastatic-malignant-pleural
- FDA approves pembrolizumab with chemotherapy for primary advanced or recurrent endometrial carcinoma, accessed October 23, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-chemotherapy-primary-advanced-or-recurrent-endometrial-carcinoma
- keytruda_pi.pdf - Merck.com, accessed October 23, 2025, https://www.merck.com/product/usa/pi_circulars/k/keytruda/keytruda_pi.pdf
- Nivolumab - NCI - Division of Cancer Treatment and Diagnosis, accessed October 23, 2025, https://dctd.cancer.gov/drug-discovery-development/reagents-materials/formulary/about/agents/nivolumab
- nivolumab - Cancer Care Ontario, accessed October 23, 2025, https://www.cancercareontario.ca/en/system/files_force/nivolumab.pdf
- Nivolumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 23, 2025, https://go.drugbank.com/drugs/DB09035
- CONSUMER MEDIA RELEASE OPDIVO (nivolumab) APPROVED IN AUSTRALIA FOR TWO CANCERS WITH THREE USES - Bristol Myers Squibb, accessed October 23, 2025, https://www.bms.com/assets/bms/australia/documents/press-releases/OPDIVO%20consumer%20media%20release.pdf
- OPDIVO (nivolumab) injection Label - accessdata.fda.gov, accessed October 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125554lbl.pdf
- OPDIVO (nivolumab) injection Label - accessdata.fda.gov, accessed October 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125554s112lbl.pdf
- Opdivo (nivolumab) dosing, indications, interactions, adverse effects, and more, accessed October 23, 2025, https://reference.medscape.com/drug/opdivo-nivolumab-999989
- Office of Clinical Pharmacology Review - FDA, accessed October 23, 2025, https://www.fda.gov/media/129248/download
- FDA-approved indications for OPDIVO® (nivolumab) and OPDIVO in combination with other therapeutic agents, accessed October 23, 2025, https://www.ajmc.com/ipubs/checkmate-77t-in-peri-operative-nsclc/FDA_Approved_Indications_062424.pdf
- Dosing Information | OPDIVO® (nivolumab), accessed October 23, 2025, https://www.opdivohcp.com/dosing/all
- Nivolumab - StatPearls - NCBI Bookshelf, accessed October 23, 2025, https://www.ncbi.nlm.nih.gov/books/NBK567801/
- Nivolumab - Wikipedia, accessed October 23, 2025, https://en.wikipedia.org/wiki/Nivolumab
- 125554Orig1s000 - accessdata.fda.gov, accessed October 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/125554orig1s000clinpharmr.pdf
- Opdivo (Nivolumab): Second PD-1 Inhibitor Receives FDA Approval for Unresectable or Metastatic Melanoma - PMC - NIH, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4665056/
- opdivo-epar-product-information_en.pdf - European Medicines Agency, accessed October 23, 2025, https://www.ema.europa.eu/en/documents/product-information/opdivo-epar-product-information_en.pdf
- Attachment: Product Information for Nivolumab - Therapeutic Goods Administration (TGA), accessed October 23, 2025, https://www.tga.gov.au/sites/default/files/auspar-nivolumab-200124-pi.pdf
- FDA approves neoadjuvant/adjuvant nivolumab for resectable non-small cell lung cancer, accessed October 23, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-neoadjuvantadjuvant-nivolumab-resectable-non-small-cell-lung-cancer
- FDA approves nivolumab in combination with cisplatin and gemcitabine for unresectable or metastatic urothelial carcinoma, accessed October 23, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-nivolumab-combination-cisplatin-and-gemcitabine-unresectable-or-metastatic-urothelial
- Libtayo® (cemiplimab-rwlc) Approved in the U.S. as First and Only Immunotherapy for Adjuvant Treatment of Cutaneous Squamous Cell Carcinoma (CSCC) with a High Risk of Recurrence After Surgery and Radiation, accessed October 23, 2025, https://newsroom.regeneron.com/news-releases/news-release-details/libtayor-cemiplimab-rwlc-approved-us-first-and-only
- Cemiplimab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 23, 2025, https://go.drugbank.com/drugs/DB14707
- www.cancercareontario.ca, accessed October 23, 2025, https://www.cancercareontario.ca/en/drugformulary/drugs/monograph/67911#:~:text=Cemiplimab%20is%20a%20recombinant%20human,1%20mediated%20immune%20response%20inhibition.
- cemiplimab - Cancer Care Ontario, accessed October 23, 2025, https://www.cancercareontario.ca/en/drugformulary/drugs/monograph/67911
- Libtayo (cemiplimab) dosing, indications, interactions, adverse effects, and more, accessed October 23, 2025, https://reference.medscape.com/drug/libtayo-cemiplimab-1000263
- LIBTAYO® (cemiplimab-rwlc) Dosing and Administration, accessed October 23, 2025, https://www.libtayohcp.com/dosing
- What is the dosing schedule for cemiplimab (Libtayo)? - Dr.Oracle, accessed October 23, 2025, https://www.droracle.ai/articles/180520/what-is-the-dosing-schedule-for-cemiplimab-libtayo
- LIBTAYO® (cemiplimab-rwlc) injection, for intravenous use - accessdata.fda.gov, accessed October 23, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/761097s014lbl.pdf
- cemiplimab | Cancer Care Ontario, accessed October 23, 2025, https://www.cancercareontario.ca/en/system/files_force/cemiplimab.pdf?download=1
- Population pharmacokinetic characteristics of cemiplimab in patients ..., accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8225544/
- Population pharmacokinetics modeling and exposure-response analyses of cemiplimab in patients with recurrent or metastatic cervical cancer - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/36251220/
- Cemiplimab - Wikipedia, accessed October 23, 2025, https://en.wikipedia.org/wiki/Cemiplimab
- FDA approves cemiplimab-rwlc for adjuvant treatment of cutaneous squamous cell carcinoma, accessed October 23, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-cemiplimab-rwlc-adjuvant-treatment-cutaneous-squamous-cell-carcinoma
- Libtayo | European Medicines Agency (EMA), accessed October 23, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/libtayo
- Libtayo - Healthdirect, accessed October 23, 2025, https://www.healthdirect.gov.au/medicines/brand/amt,1506831000168109/libtayo
- Pembrolizumab Utilization and Clinical Outcomes Among Patients ..., accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8191741/
- keytruda is an fda-approved treatment for advanced melanoma, accessed October 23, 2025, https://www.keytruda.com/melanoma/advanced/
- Investigating the Long-Term Effects of Nivolumab and Ipilimumab in ..., accessed October 23, 2025, https://www.nyp.org/advances/article/cancer/investigating-the-long-term-effects-of-nivolumab-and-ipilimumab-in-advanced-melanoma
- Five-Year Survival Data for Merck's KEYTRUDA® (pembrolizumab ..., accessed October 23, 2025, https://www.merck.com/news/five-year-survival-data-for-mercks-keytruda-pembrolizumab-in-advanced-non-small-cell-lung-cancer-nsclc-from-first-keynote-trial-at-2019-asco-annual-meeting/
- Breakthrough 5-year survival with pembrolizumab in Keynote-001 study: horizon shifting in advanced non-small cell lung cancer with immune check point inhibition - de Silva - Annals of Translational Medicine, accessed October 23, 2025, https://atm.amegroups.org/article/view/36943/html
- KEYTRUDA® (pembrolizumab) Demonstrates Long-Term Survival Benefit in Certain Patients With Earlier or Advanced Stages of Non-Small Cell Lung Cancer (NSCLC), accessed October 23, 2025, https://www.merck.com/news/keytruda-pembrolizumab-demonstrates-long-term-survival-benefit-in-certain-patients-with-earlier-or-advanced-stages-of-non-small-cell-lung-cancer-nsclc/
- Five-year outcomes from the randomized, phase iii trials checkmate ..., accessed October 23, 2025, https://profiles.foxchase.org/en/publications/five-year-outcomes-from-the-randomized-phase-iii-trials-checkmate
- Five-Year Outcomes From the CheckMate 017 and 057 Trials of Nivolumab vs Docetaxel in Previously Treated Patients With NSCLC - The ASCO Post, accessed October 23, 2025, https://ascopost.com/news/february-2021/five-year-outcomes-from-the-checkmate-017-and-057-trials-of-nivolumab-vs-docetaxel-in-previously-treated-patients-with-nsclc/
- Sustained Survival Shown in Updated Nivolumab Results in NSCLC | Targeted Oncology - Immunotherapy, Biomarkers, and Cancer Pathways, accessed October 23, 2025, https://www.targetedonc.com/view/sustained-survival-shown-in-updated-nivolumab-results-in-nsclc
- Nivolumab Versus Docetaxel in Previously Treated Patients With Advanced Non-Small-Cell Lung Cancer: Two-Year Outcomes From Two Randomized, Open-Label, Phase III Trials (CheckMate 017 and CheckMate 057) - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/29023213/
- Cemiplimab Potential New Treatment Option for Patients with Advanced NSCLC and PD-L1 ≥50% - The Oncology Pharmacist, accessed October 23, 2025, https://theoncologypharmacist.com/articles/cemiplimab-potential-new-treatment-option-for-patients-with-advanced-nsclc-and-pd-l1-50
- Libtayo® (cemiplimab) Plus Chemotherapy Results at Five Years ..., accessed October 23, 2025, https://newsroom.regeneron.com/news-releases/news-release-details/libtayor-cemiplimab-plus-chemotherapy-results-five-years/
- Confirmed safety and efficacy of cemiplimab in patients with advanced cutaneous squamous cell carcinoma - BJMO, accessed October 23, 2025, https://www.bjmo.be/confirmed-safety-and-efficacy-of-cemiplimab-in-patients-with-advanced-cutaneous-squamous-cell-carcinoma/
- Longer Follow-Up for Cemiplimab Confirms Superior Efficacy Over ..., accessed October 23, 2025, https://www.onclive.com/view/longer-followup-for-cemiplimab-confirms-superior-efficacy-over-other-regimens-for-cscc
- A phase 2 open-label study of cemiplimab in patients with advanced cutaneous squamous cell carcinoma (EMPOWER-CSCC-1): Final long-term analysis of groups 1, 2, and 3, and primary analysis of fixed-dose treatment group 6 - WashU Medicine Research Profiles, accessed October 23, 2025, https://profiles.wustl.edu/en/publications/a-phase-2-open-label-study-of-cemiplimab-in-patients-with-advance
- KEYTRUDA® (pembrolizumab) Showed Statistically Significant Improvement In Disease-Free Survival Versus Placebo As Adjuvant Treatment For Patients With Stage IB-IIIA Non-Small Cell Lung Cancer Regardless Of PD-L1 Expression - EORTC, accessed October 23, 2025, https://www.eortc.org/blog/2022/01/10/keytruda-pembrolizumab-showed-statistically-significant-improvement-in-disease-free-survival-as-adjuvant-treatment/
- Comparative Analysis of Predictive Biomarkers for PD-1/PD-L1 Inhibitors in Cancers: Developments and Challenges - MDPI, accessed October 23, 2025, https://www.mdpi.com/2072-6694/14/1/109
- Rationale for PD-L1 Expression as a Biomarker in Immuno-Oncology, accessed October 23, 2025, https://www.theoncologynurse.com/articles/rationale-for-pd-l1-expression-as-a-biomarker-in-immuno-oncology
- Attachment-Product information for Keytruda - Therapeutic Goods Administration (TGA), accessed October 23, 2025, https://www.tga.gov.au/sites/default/files/2023-06/auspar-keytruda-230608-pi.pdf
- Companion diagnostics and predictive biomarkers for PD-1/PD-L1 immune checkpoint inhibitors therapy in malignant melanoma - Frontiers, accessed October 23, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1454720/full
- Research progress of biomarkers in the prediction of anti-PD-1/PD-L1 immunotherapeutic efficiency in lung cancer - PMC, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10351040/
- Case Study: PD-L1 and Immunotherapy Response, Biomarker Research, accessed October 23, 2025, https://www.curemelanoma.org/case-studies/predicting-response-to-immunotherapy-pd-l1-as-a-biomarker-and-beyond
- Evaluating Tumour Mutational Burden as a Key Biomarker in Personalized Cancer Immunotherapy: A Pan-Cancer Systematic Review - MDPI, accessed October 23, 2025, https://www.mdpi.com/2072-6694/17/3/480
- Study finds high tumor mutation burden predicts immunotherapy response in some, but not all, cancers, accessed October 23, 2025, https://www.mdanderson.org/newsroom/tumor-mutation-burden-predicts-immunotherapy-response-some-not-all-cancers.h00-159459267.html
- Evaluating Immune Checkpoint Inhibition Biomarkers - The Jackson Laboratory, accessed October 23, 2025, https://www.jax.org/education-and-learning/clinical-and-continuing-education/clinical-topics/tumor-testing/pd-l1
- Biomarkers for Response to Anti-PD-1/Anti-PD-L1 Immune Checkpoint Inhibitors: A Large Meta-Analysis - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/37216635/
- Cemiplimab - DermNet, accessed October 23, 2025, https://dermnetnz.org/topics/cemiplimab
- Safety profile of pembrolizumab monotherapy based on an aggregate safety evaluation of 8937 patients - NIH, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11227881/
- Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis - NIH, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6487913/
- Immune-related adverse events in patients with gastrointestinal cancer undergoing pembrolizumab monotherapy: A systematic review of clinical trials. - ASCO Publications, accessed October 23, 2025, https://ascopubs.org/doi/10.1200/JCO.2024.42.3_suppl.744
- Safety profile - LIBTAYO® (cemiplimab-rwlc), accessed October 23, 2025, https://www.libtayohcp.com/cscc/safety
- 4151-Locally advanced or metastatic cemiplimab - eviQ, accessed October 23, 2025, https://www.eviq.org.au/medical-oncology/respiratory/non-small-cell-lung-cancer-advanced-metastatic/4151-locally-advanced-or-metastatic-cemiplimab
- Immune Checkpoint Inhibitor Toxicity Management - Clinical Practice Guideline - Cancer Care Ontario, accessed October 23, 2025, https://www.cancercareontario.ca/sites/ccocancercare/files/guidelines/full/ImmuneCheckpointInhibitor.pdf
- Anti-PD-1 and Novel Combinations in the Treatment of Melanoma ..., accessed October 23, 2025, https://www.mdpi.com/2077-0383/9/1/223
- Risk of Adverse Events in Cancer Patients Receiving Nivolumab With Ipilimumab: A Meta-Analysis - Frontiers, accessed October 23, 2025, https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2022.877434/full
- Antibody therapeutics approved or in regulatory review in the EU or US, accessed October 23, 2025, https://www.antibodysociety.org/resources/approved-antibodies/
- Merck Receives Two Positive EU CHMP Opinions for KEYTRUDA® (pembrolizumab), for Subcutaneous (SC) Administration and for New Indication for Earlier-Stage Head and Neck Cancer, accessed October 23, 2025, https://www.merck.com/news/merck-receives-two-positive-eu-chmp-opinions-for-keytruda-pembrolizumab-for-subcutaneous-sc-administration-and-for-new-indication-for-earlier-stage-head-and-neck-cancer/
- Pharmacokinetics and safety of subcutaneous nivolumab: results from the phase I/II CheckMate 8KX study - The Journal for ImmunoTherapy of Cancer, accessed October 23, 2025, https://jitc.bmj.com/content/13/10/e011918.full.pdf
- Targeting PD-1/PD-L-1 immune checkpoint inhibition for cancer immunotherapy: success and challenges - Frontiers, accessed October 23, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1383456/full