2022215-59-2
Advanced Microsatellite Instability High Endometrial Carcinoma, Advanced Mismatch Repair-deficient (dMMR) Endometrial Cancer, Advanced Mismatch repair deficient (dMMR) solid tumors, Recurrent Microsatellite Instability (MSI)-High Endometrial Cancer, Recurrent Mismatch Repair-deficient (dMMR) Endometrial Cancer, Recurrent Mismatch repair deficient (dMMR) solid tumors
Dostarlimab, marketed under the brand name Jemperli, is a humanized immunoglobulin G4 (IgG4) monoclonal antibody that has emerged as a pivotal therapeutic agent in the field of immuno-oncology. As a programmed death receptor-1 (PD-1) blocking antibody, dostarlimab functions by reinvigorating the host's immune system to combat malignant cells. Its mechanism hinges on high-affinity binding to the PD-1 receptor on T-cells, thereby preventing its interaction with ligands PD-L1 and PD-L2 and releasing the "brake" on anti-tumor immunity.
The clinical development of dostarlimab has been marked by rapid and significant success, particularly in cancers characterized by a deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H) biomarker status. Landmark clinical trials have established dostarlimab as a new standard of care in endometrial cancer (EC). The GARNET study demonstrated robust and exceptionally durable responses for dostarlimab monotherapy in patients with previously treated, advanced dMMR EC. Subsequently, the RUBY trial established its role in the first-line setting, where its addition to chemotherapy conferred a profound survival benefit in patients with dMMR/MSI-H primary advanced or recurrent EC and also improved overall survival in the broader all-comer population, leading to expanded global approvals.
Beyond endometrial cancer, dostarlimab has produced unprecedented results in a phase II study for locally advanced dMMR rectal cancer, achieving a 100% clinical complete response rate and heralding a potential paradigm shift toward non-operative, curative-intent management. This has been complemented by a tumor-agnostic accelerated approval for the treatment of any dMMR solid tumor that has progressed on prior therapy.
The safety profile of dostarlimab is consistent with its drug class, characterized primarily by immune-mediated adverse reactions (IMARs) that can affect any organ system. These reactions, while potentially severe, are generally manageable with established protocols involving corticosteroids. The pharmacokinetic profile, notable for a long elimination half-life of approximately 25 days, supports a convenient every-six-week maintenance dosing schedule, reducing the treatment burden for patients.
This report provides a comprehensive analysis of dostarlimab, synthesizing data on its pharmacology, pivotal clinical trial results, safety profile, regulatory history, and comparative positioning. It aims to serve as an exhaustive scientific and clinical monograph for this important antineoplastic agent.
Dostarlimab is a biologic therapeutic agent classified as a protein-based therapy.[1] It is a humanized monoclonal antibody of the IgG4 kappa isotype, produced using recombinant DNA technology in a mammalian cell line (Chinese Hamster Ovary, CHO).[2] In the United States, its non-proprietary name is often listed as dostarlimab-gxly, a convention used for certain biologics to distinguish them from potential future biosimilars.[5] The drug was developed under several internal codes, including TSR-042, GSK-4057190A, and ANB011.[8] Its fundamental properties are summarized in Table 2.1.
Table 2.1: Key Drug Identification and Properties
Property | Detail | Source(s) |
---|---|---|
Non-proprietary Name | Dostarlimab, Dostarlimab-gxly | 5 |
Proprietary Name | Jemperli® | 9 |
DrugBank Accession Number | DB15627 | 1 |
CAS Registry Number | 2022215-59-2 | 10 |
Drug Class | Anti-PD-1 Monoclonal Antibody, Immune Checkpoint Inhibitor, Antineoplastic Agent | 6 |
Molecular Type | Humanized IgG4 kappa Monoclonal Antibody | 1 |
Manufacturing | Recombinant DNA technology in Chinese Hamster Ovary (CHO) cells | 3 |
The therapeutic activity of dostarlimab is rooted in its function as an immune checkpoint inhibitor.[2] The core of its mechanism is the specific and high-affinity binding to the PD-1 receptor, which is expressed on the surface of activated T-cells.[13] In a healthy immune system, the PD-1 pathway acts as a crucial negative feedback loop to maintain self-tolerance and modulate the intensity and duration of immune responses, preventing autoimmunity.[14] Some cancer cells exploit this natural regulatory mechanism to evade immune destruction. They achieve this by overexpressing the ligands for PD-1, namely Programmed Death-Ligand 1 (PD-L1) and Programmed Death-Ligand 2 (PD-L2).[2] When these ligands bind to the PD-1 receptor on T-cells, they deliver a potent inhibitory signal that leads to T-cell exhaustion and apoptosis, effectively creating an immunosuppressive tumor microenvironment.[14]
Dostarlimab acts by physically obstructing this interaction. It binds to the PD-1 receptor and competitively inhibits its engagement with both PD-L1 and PD-L2.[16] This blockade removes the suppressive signal, thereby "releasing the brake" on T-cell activity.[14] The result is the restoration of T-cell proliferation, cytokine production (such as Interleukin-2 and Interferon-gamma), and cytotoxic T-lymphocyte (CTL) activity, enabling the immune system to once again recognize and mount an effective attack against tumor cells.[13]
The efficacy of this blockade is underpinned by dostarlimab's exceptional binding affinity. Laboratory studies have quantified this potency, showing half-maximal inhibitory concentrations (IC50) of 1.8 nM for blocking the PD-1/PD-L1 interaction and 1.5 nM for the PD-1/PD-L2 interaction.[20] Furthermore, its equilibrium dissociation constant (
Kd), a measure of binding strength, for human PD-1 is approximately 0.3 nM.[8] These sub-nanomolar and low-nanomolar values signify an extremely tight and durable bond between dostarlimab and the PD-1 receptor. This potent binding allows dostarlimab to effectively outcompete the natural ligands, ensuring a near-complete and sustained blockade of the checkpoint pathway. This robust molecular activity provides a direct biological rationale for the durable clinical responses observed in pivotal trials, where many patients experience long-term disease control.[7]
The pharmacodynamic effects of dostarlimab confirm that its high-affinity molecular binding translates into a functional biological response. In vitro cellular assays have demonstrated that dostarlimab treatment leads to potent T-cell activation. In a human CD4+ T-cell mixed lymphocyte reaction (MLR) assay, dostarlimab increased Interleukin-2 (IL-2) production with a half-maximal effective concentration (EC50) of approximately 1 nM.[20] In staphylococcal enterotoxin B (SEB)-stimulated peripheral blood mononuclear cells (PBMCs), the
EC50 for IL-2 production was even lower, at approximately 0.1 nM.[8] Furthermore, dostarlimab enhances antigen-specific T-cell activation, as shown by an increase in interferon-gamma (IFN-γ) release with an
EC50 of approximately 0.5 nM.[15] Importantly, dostarlimab does not elicit non-specific T-cell responses in the absence of an antigen, confirming that it functions as a true checkpoint blocker rather than a non-specific T-cell agonist.[15]
This cellular activity has been shown to alter the tumor microenvironment in a manner favorable to anti-tumor immunity. In preclinical in vivo studies using an A549 lung cancer model, treatment with dostarlimab resulted in significant tumor growth inhibition of 62% at study termination compared to controls.[20] This anti-tumor activity was directly associated with pharmacodynamic changes within the tumor itself, including an increased infiltration of tumor-killing CD8+ T-cells and a concurrent reduction in the population of immunosuppressive regulatory T-cells (Tregs).[20] This chain of evidence—from high-affinity binding to functional T-cell activation and finally to a pro-inflammatory shift in the tumor microenvironment—provides a comprehensive, multi-level validation of the drug's mechanism of action prior to clinical application.
Dostarlimab is administered via intravenous (IV) infusion over a period of 30 minutes.[1] Its pharmacokinetics (PK) are linear in the dose range of 1 mg/kg to 10 mg/kg.[19] The drug's behavior in the body has been characterized in patients with various solid tumors, including endometrial cancer.[24]
Following the initial dosing regimen of 500 mg every 3 weeks, the mean maximum concentration (Cmax) is 171 mcg/mL. During the maintenance phase of 1000 mg every 6 weeks, the mean Cmax reaches 309 mcg/mL.[1] The mean volume of distribution (
Vd) at steady-state is approximately 5.3 L.[2] This relatively small volume of distribution suggests that the drug is primarily confined to the vascular and interstitial compartments, consistent with a large molecule like a monoclonal antibody.
As a protein, dostarlimab is not metabolized by cytochrome P450 enzymes or other traditional drug metabolism pathways. Instead, it is expected to be degraded into small peptides and amino acids via non-specific catabolic pathways throughout the body.[1] The mean clearance (CL) of dostarlimab at steady-state is low, at 0.007 L/h.[1] This slow clearance contributes to a long mean terminal elimination half-life (
t1/2) of 25.4 days.[1]
This extended half-life is a critical pharmacokinetic feature that directly enables the convenient maintenance dosing schedule of 1000 mg every 6 weeks (Q6W).[13] A dosing interval of 42 days is approximately 1.6 times the drug's half-life, an interval that maintains drug concentrations well within the therapeutic window. This less frequent dosing regimen represents a significant quality-of-life benefit for patients, reducing the burden of treatment by minimizing the frequency of clinic visits compared to more common three- or four-week cycles for other cancer therapies.
No clinically significant differences in the pharmacokinetics of dostarlimab have been observed based on age (24 to 86 years), sex, race, tumor type, or in patients with renal or hepatic impairment.[24] A summary of key pharmacokinetic parameters is provided in Table 2.2.
Table 2.2: Summary of Dostarlimab Pharmacokinetic Parameters
Parameter | Value | Source(s) |
---|---|---|
Administration Route | Intravenous Infusion | 23 |
Infusion Time | 30 minutes | 1 |
Volume of Distribution (Vd) | ~5.3 L | 2 |
Clearance (CL) | ~0.007 L/h | 1 |
Elimination Half-Life (t1/2) | ~25.4 days | 1 |
Metabolism | Catabolic pathways | 1 |
Dostarlimab has fundamentally altered the treatment landscape for endometrial cancer (EC), a common gynecologic malignancy that arises from the lining of the uterus.[9] Its efficacy has been proven in two pivotal clinical trials, GARNET and RUBY, leading to its establishment as a new standard of care in both previously treated and frontline settings.
The GARNET trial (NCT02715284) was a multicenter, single-arm, open-label phase I/II study that provided the foundational evidence for dostarlimab's utility in EC.[7] The trial evaluated dostarlimab monotherapy in patients with advanced solid tumors, with a specific expansion cohort (A1) dedicated to patients with dMMR/MSI-H advanced or recurrent EC whose disease had progressed on or after a platinum-containing chemotherapy regimen.[26]
In an efficacy population of 141 patients from this cohort, dostarlimab demonstrated impressive and durable anti-tumor activity.[7] Based on a blinded independent central review (BICR) with a median follow-up of 27.9 months, the key results were as follows [7]:
Efficacy was observed consistently across patient subgroups, including those who had received one prior line of therapy (ORR 43.8%) and those who were more heavily pre-treated with two or more prior lines (ORR 48.1%).[28] The most compelling finding from GARNET was not merely the response rate but the profound durability of these responses. With a median DOR not reached at over two years of follow-up, the data suggest that for the nearly half of patients who respond, dostarlimab can induce a long-term, stable disease control. This represents a significant clinical achievement, potentially transforming what was a progressively fatal disease into a chronic, manageable condition for this biomarker-defined subset of patients. These results led to the initial regulatory approvals for dostarlimab in this setting.[7]
Table 3.1: Summary of Efficacy Results from the GARNET Trial (Cohort A1)
Endpoint | Result (N=141) | 95% Confidence Interval | Source(s) |
---|---|---|---|
Objective Response Rate (ORR) | 45.5% | 37.1% - 54.0% | 7 |
Complete Response (CR) Rate | 15.6% | - | 7 |
Partial Response (PR) Rate | 29.8% | - | 7 |
Median Duration of Response (DOR) | Not Reached | - | 7 |
% Responders with DOR ≥12 months | 85.9% | - | 7 |
% Responders with DOR ≥24 months | 54.7% | - | 7 |
Building on the success of GARNET, the RUBY trial (NCT03981796) evaluated dostarlimab in the first-line setting.[9] This randomized, double-blind, placebo-controlled phase III trial compared dostarlimab plus standard-of-care chemotherapy (carboplatin and paclitaxel) against placebo plus chemotherapy in patients with primary advanced or recurrent EC.[29] The trial's design allowed for analysis in both a pre-specified dMMR/MSI-H subgroup and the overall (all-comer) population, yielding practice-changing results for both.
The magnitude of benefit demonstrated in the RUBY trial has redefined the first-line standard of care for advanced or recurrent EC. The profound benefit in the dMMR population (HR 0.29 for PFS) is so large that it effectively mandates the inclusion of dostarlimab with chemotherapy for these patients. Concurrently, the statistically significant OS benefit observed in the overall population, while heavily influenced by the dMMR subgroup, was robust enough to convince regulatory agencies like the U.S. FDA and the European Commission to expand the approval to all patients with primary advanced or recurrent EC, regardless of their MMR status.[32] This has fundamentally changed the treatment algorithm for a much larger group of patients than initially targeted.
Table 3.2: Key Efficacy Outcomes from the RUBY Trial (dMMR/MSI-H and Overall Populations)
Population | Endpoint | Dostarlimab + Chemo Arm | Placebo + Chemo Arm | Hazard Ratio (95% CI) | p-value | Source(s) |
---|---|---|---|---|---|---|
dMMR/MSI-H | Median PFS | 30.3 months | 7.7 months | 0.29 (0.17, 0.50) | <0.0001 | 29 |
Overall | Median OS | 44.6 months | 28.2 months | 0.69 (0.54, 0.89) | 0.002 | 30 |
Dostarlimab has generated unprecedented results in a population outside of its primary indication. A phase II investigator-initiated trial conducted at Memorial Sloan Kettering Cancer Center (MSK) evaluated dostarlimab monotherapy as a neoadjuvant (pre-surgical) treatment for patients with locally advanced dMMR rectal cancer.[22] The standard of care for this disease typically involves a grueling regimen of chemotherapy, radiation, and major surgery, often resulting in permanent colostomy and significant long-term morbidity.[35]
The results from this trial were remarkable. An initial report on the first 12 patients, published in The New England Journal of Medicine, showed a 100% clinical complete response (cCR) rate.[2] Updated, longer-term data presented at the 2024 ASCO Annual Meeting confirmed and extended this finding, showing a 100% cCR rate in all 42 patients who had completed treatment.[22] This response was defined as no evidence of tumor on magnetic resonance imaging (MRI), endoscopy, and digital rectal exam.[22]
Crucially, every patient who achieved a cCR was able to forgo subsequent chemoradiation and surgery.[6] The responses have proven to be durable, with a median follow-up of 26.3 months in the first 24 patients, and some responses have been sustained for as long as five years.[22] These findings represent more than just high efficacy; they signal a potential paradigm shift toward non-operative management and organ preservation. If these results can be replicated in larger, ongoing registrational trials such as AZUR-1 and AZUR-2, dostarlimab could establish a new curative-intent treatment pathway that eliminates the need for life-altering surgery and toxic radiation for this patient subset, representing a transformative improvement in survivorship and quality of life.[22]
Reflecting a broader shift in oncology toward biomarker-driven therapy, dostarlimab received accelerated approval from the FDA for a tumor-agnostic indication.[9] This approval covers the treatment of adult patients with dMMR recurrent or advanced solid tumors, as determined by an FDA-approved test, that have progressed on or following prior treatment and who have no satisfactory alternative treatment options.[1]
This approval was based on data from the GARNET study, which included a cohort (F) for non-endometrial dMMR solid tumors.[27] This pan-tumor indication underscores the principle that the genetic biomarker of dMMR/MSI-H is a more powerful predictor of response to PD-1 blockade than the anatomical site of the cancer's origin. Tumors with this biomarker have a defective DNA repair mechanism, leading to a very high number of mutations and the generation of tumor neoantigens.[15] These neoantigens make the cancer cells highly "visible" to a reawakened immune system, explaining the high response rates to checkpoint inhibitors across different tumor types, including gastric and colorectal cancers.[38] This approval emphasizes the critical importance of universal biomarker testing for all patients with advanced cancer to identify those who may be candidates for this highly effective therapy.[35] As an accelerated approval, continued approval for this indication is contingent upon verification of clinical benefit in confirmatory trials.[1]
The clinical development program for dostarlimab is extensive and continues to expand, aiming to build on its established successes and explore new frontiers.[40] A primary focus is on confirming the groundbreaking results in rectal cancer through larger, registrational studies. The AZUR-1 trial is investigating dostarlimab monotherapy as a replacement for standard neoadjuvant therapy in dMMR/MSI-H locally advanced rectal cancer, while the AZUR-2 trial is evaluating perioperative dostarlimab in resectable dMMR/MSI-H colon cancer.[22]
Beyond colorectal cancer, dostarlimab is being actively studied in a wide range of other malignancies. Active phase II trials are recruiting patients with gastric adenocarcinoma [38], and other studies are exploring its use in non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma, and melanoma.[40]
A significant portion of the investigational strategy involves evaluating dostarlimab in rational combination regimens designed to overcome resistance and enhance efficacy. Trials are underway combining dostarlimab with other immunotherapies, such as the TIM-3 inhibitor cobolimab, based on the rationale that dual checkpoint blockade may be synergistic.[40] Another key area is the combination with targeted therapies, such as PARP inhibitors (niraparib) in cervical and penile cancer.[40] The scientific basis for this approach is that PARP inhibitors can induce DNA damage and genomic instability, potentially increasing the immunogenicity of tumors and making them more susceptible to PD-1 blockade. This broad and scientifically driven clinical trial program demonstrates a clear strategy: first, to solidify and expand its use in dMMR/MSI-H tumors, and second, to push the boundaries of immunotherapy by exploring novel combinations to tackle more challenging and resistant cancers.
As with all immune checkpoint inhibitors, the therapeutic benefit of dostarlimab is accompanied by a unique spectrum of potential adverse effects, primarily immune-mediated adverse reactions (IMARs). Understanding and managing this safety profile is paramount for its safe and effective use.
The mechanism of action of dostarlimab—releasing the brakes on the immune system—inherently carries the risk of inducing an overactive immune response that can target healthy tissues.[1] These IMARs can be severe or fatal, can occur at any time during or after treatment, and can affect any organ system or tissue.[1]
Prompt recognition and management are critical. The general management principle is based on the severity of the reaction, graded according to the National Cancer Institute's Common Terminology Criteria for Adverse Events (CTCAE). For suspected IMARs, dostarlimab should be withheld, and appropriate workup should be initiated to exclude other causes, such as infection.[6] For moderate (Grade 2) reactions, treatment typically involves withholding dostarlimab and initiating systemic corticosteroids (e.g., prednisone 1 to 2 mg/kg/day or equivalent). For severe (Grade 3) or life-threatening (Grade 4) reactions, dostarlimab should be permanently discontinued, and high-dose corticosteroids should be administered.[23] Once the reaction improves to Grade 1 or less, the corticosteroid dose should be tapered slowly over at least one month to prevent recurrence.[6] Specific dose modification recommendations are outlined in Table 4.1.
Table 4.1: Recommended Dose Modifications for Immune-Mediated Adverse Reactions
IMAR Type | Grade 2 Severity | Grade 3 or 4 Severity | Source(s) |
---|---|---|---|
Pneumonitis | Withhold | Permanently discontinue (or for recurrent Grade 2) | 23 |
Colitis | Withhold | Withhold (Grade 3), Permanently discontinue (Grade 4) | 3 |
Hepatitis | Withhold | Permanently discontinue | 3 |
Endocrinopathies | Withhold until clinically stable | Withhold or permanently discontinue depending on severity | 3 |
Nephritis with renal dysfunction | Withhold | Permanently discontinue | 23 |
Exfoliative dermatologic conditions | Withhold (suspected SJS/TEN) | Permanently discontinue (confirmed SJS/TEN) | 23 |
Myocarditis | Permanently discontinue | Permanently discontinue | 23 |
Neurological toxicities | Withhold | Permanently discontinue | 23 |
Several specific IMARs have been reported in clinical trials with dostarlimab:
Beyond the specific category of IMARs, dostarlimab is associated with a profile of more general adverse events (AEs). The profile differs depending on whether it is used as a single agent or in combination with chemotherapy. The overlap in toxicities between dostarlimab and chemotherapy creates a clinical challenge in correctly attributing symptoms. For example, a patient on combination therapy presenting with diarrhea requires careful evaluation to differentiate between a common chemotherapy side effect, which can be managed supportively, and potentially life-threatening immune-mediated colitis, which requires immediate drug cessation and immunosuppression.
The regulatory history of dostarlimab illustrates a rapid and successful development pathway, progressing from a niche indication to a broad standard of care in a remarkably short timeframe. This trajectory was enabled by well-designed pivotal trials that demonstrated compelling efficacy in areas of high unmet medical need.
This step-wise expansion, moving from an accelerated approval in a biomarker-defined subgroup to a full approval as a broad first-line standard of care in just over three years, serves as a model for efficient and impactful drug development.
While dostarlimab belongs to the same class as other established PD-1 inhibitors like pembrolizumab and nivolumab, emerging data suggest potential differences in binding, efficacy, and approved uses.
Table 5.1: Comparative Efficacy and Safety of Dostarlimab vs. Pembrolizumab in NSCLC (PERLA Trial)
Endpoint | Dostarlimab + Chemo Arm | Pembrolizumab + Chemo Arm | Hazard Ratio / Difference (95% CI) | Source(s) |
---|---|---|---|---|
Median PFS | 8.8 months | 6.7 months | HR 0.70 (0.50, 0.98) | 41 |
Median OS | 19.4 months | 15.9 months | HR 0.75 (0.53, 1.05) | 41 |
ORR | 46% | 37% | Difference 9% | 45 |
Grade ≥3 TRAEs | 36% | 42% | - | 45 |
Serious AEs | 38% | 45% | - | 45 |
The significant clinical benefits of dostarlimab are accompanied by high acquisition costs, leading to complex health economic assessments that vary by patient population and healthcare system.
A cost-effectiveness analysis from the perspective of a United States payer found that dostarlimab in combination with chemotherapy is a cost-effective option for the first-line treatment of primary advanced or recurrent dMMR EC. The analysis yielded an incremental cost-effectiveness ratio (ICER) of approximately $60,349 per quality-adjusted life year (QALY) gained, a value that falls comfortably below the common willingness-to-pay threshold of $150,000 per QALY.[53]
However, for the pMMR EC population, the same analysis found that the combination was not cost-effective, with an ICER of approximately $175,788 per QALY.[53] This highlights a significant tension between regulatory approval and economic value. While the drug is approved for the all-comer population based on a statistically significant OS benefit, the magnitude of that benefit is smaller in the pMMR subgroup than in the dMMR subgroup. This results in a high cost per unit of clinical benefit gained, which may lead to reimbursement challenges and access hurdles for pMMR patients in certain healthcare systems.
The economic picture also varies globally. A similar analysis from the perspective of the Chinese healthcare system concluded that dostarlimab plus chemotherapy was unlikely to be a cost-effective option for advanced EC in either the dMMR or pMMR subgroups, given the local willingness-to-pay thresholds.[54]
Dostarlimab is classified as a "biotech" drug, meaning it is produced through a complex biological process rather than by traditional chemical synthesis.[55] This manufacturing process is intricate, multi-staged, and requires stringent control to ensure the final product's quality, safety, and efficacy.[55]
This complex biological manufacturing process is a key reason for the high cost of monoclonal antibodies like dostarlimab. It also underscores the importance of proper storage and handling (e.g., refrigeration, use of specific infusion materials) to maintain the protein's structural integrity and biological activity.[10] Furthermore, because the final product is a large protein, there is an inherent risk of immunogenicity, where the patient's body may develop anti-drug antibodies (ADAs), which have the potential to affect the drug's pharmacokinetics and efficacy.[6]
Dostarlimab has firmly established itself as a major therapeutic advance in oncology, exemplifying the power of targeted immunotherapy. Its development showcases a successful translation of potent molecular pharmacology—characterized by high-affinity and durable PD-1 blockade—into profound and practice-changing clinical benefit. In the landscape of dMMR/MSI-H cancers, it has become a cornerstone therapy. For patients with advanced endometrial cancer, it has redefined the standard of care in both second-line and, most critically, first-line settings. For those with locally advanced rectal cancer, it has offered the unprecedented potential for a cure without the need for debilitating surgery or radiation.
The rapid regulatory trajectory of dostarlimab, moving from a niche accelerated approval to a broad, all-comer indication in a major cancer type, highlights its significant impact and the strength of the clinical data supporting its use. Furthermore, head-to-head data from the PERLA trial suggest it may have a competitive efficacy profile against the established market leader, pembrolizumab, in NSCLC, though further confirmation is needed.
Despite these successes, the story of dostarlimab is still unfolding, and several key questions will shape its future role in cancer therapy:
In conclusion, dostarlimab (Jemperli) is a powerful and effective PD-1 inhibitor that has already transformed the lives of patients with specific types of cancer. Its journey from a targeted agent for a niche biomarker to a broad standard of care is a testament to its efficacy and the success of its clinical development program. The ongoing research will continue to define its ultimate place in the armamentarium of cancer therapies, with the potential to bring durable responses and improved quality of life to an even wider range of patients.
Published at: July 30, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.