Sasanlimab (PF-06801591) is an investigational, humanized immunoglobulin G4 (IgG4) monoclonal antibody developed by Pfizer that functions as an immune checkpoint inhibitor by targeting the programmed cell death 1 (PD-1) receptor. Its mechanism involves blocking the interaction between PD-1 and its ligands, PD-L1 and PD-L2, thereby restoring the anti-tumor activity of the host immune system. The most significant clinical advancement for sasanlimab has been in the treatment of high-risk, Bacillus Calmette-Guérin (BCG)-naïve non-muscle invasive bladder cancer (NMIBC). In the pivotal, multinational Phase III CREST trial, the combination of subcutaneously administered sasanlimab with standard-of-care intravesical BCG (induction and maintenance) demonstrated a statistically significant and clinically meaningful improvement in the primary endpoint of event-free survival (EFS) compared to BCG alone. The combination therapy reduced the risk of an EFS event by 32% and showed particularly durable complete responses in patients with carcinoma in situ (CIS).
Sasanlimab is distinguished from other approved immunotherapies in urothelial cancer by two key strategic features. First, it is being developed for the BCG-naïve patient population, an earlier treatment setting than the BCG-unresponsive indication for which pembrolizumab is approved, thus carving out a distinct therapeutic niche. Second, its formulation for subcutaneous administration offers significant logistical and practical advantages over the intravenous infusions required for many other checkpoint inhibitors, potentially lowering the treatment burden for patients and healthcare systems and enabling administration in a wider range of clinical settings, including urology practices. The safety profile of the sasanlimab-BCG combination is manageable and consistent with the known toxicities of each agent individually, although the addition of systemic immunotherapy leads to a higher incidence of serious and immune-related adverse events compared to BCG monotherapy. Pending regulatory review of the robust data from the CREST trial, sasanlimab in combination with BCG has the potential to become the first new standard of care for high-risk, BCG-naïve NMIBC in more than three decades.
Sasanlimab, also known by its developmental code PF-06801591, is an investigational therapeutic agent developed by the pharmaceutical company Pfizer. It is classified as a humanized immunoglobulin G4-kappa (IgG4-κ) monoclonal antibody and belongs to the rapidly expanding class of cancer treatments known as immune checkpoint inhibitors (ICIs). The specific molecular target of sasanlimab is the programmed cell death 1 (PD-1) receptor, a critical negative regulator of the adaptive immune response. Through its targeted action, sasanlimab is being evaluated for its potential antineoplastic activities across a spectrum of malignancies, with its most advanced clinical development focused on urothelial carcinoma, particularly non-muscle invasive bladder cancer (NMIBC).
The therapeutic principle of sasanlimab is rooted in the complex biology of the PD-1/PD-L1 immune checkpoint pathway. PD-1 is an inhibitory co-stimulatory receptor that is expressed on the surface of activated immune cells, including T lymphocytes, B lymphocytes, natural killer (NK) cells, dendritic cells, and macrophages. This receptor plays a crucial role in maintaining peripheral tolerance and preventing excessive or prolonged immune responses, thereby protecting host tissues from autoimmune damage.
The function of PD-1 is mediated through its interaction with two primary ligands: programmed death-ligand 1 (PD-L1) and programmed death-ligand 2 (PD-L2). When PD-1 on an activated T cell binds to PD-L1 or PD-L2 on another cell, it initiates a cascade of downstream intracellular signaling events that effectively suppress T-cell activation, proliferation, cytokine production, and cytotoxic effector functions. Many types of cancer cells have co-opted this natural regulatory mechanism to evade destruction by the immune system. Tumor cells frequently upregulate the expression of PD-L1 on their surface, allowing them to engage with PD-1 on tumor-infiltrating T cells and effectively "turn off" the anti-tumor immune attack. This interaction is a primary mechanism of tumor immune evasion and is fundamental to the progression of many malignancies.
The therapeutic strategy of PD-1 blockade is to disrupt this tumor-driven immunosuppression. By binding with high affinity to the PD-1 receptor, an antagonistic antibody like sasanlimab physically blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This action effectively "releases the brakes" on the immune system, preventing the inhibitory signaling and restoring the ability of cytotoxic T lymphocytes (CTLs) and other effector immune cells to recognize and eliminate cancer cells.
In the context of NMIBC, there is a particularly compelling synergistic rationale for combining a PD-1 inhibitor with the long-established standard of care, intravesical Bacillus Calmette-Guérin (BCG) therapy. BCG is a live attenuated bacterium that, when instilled into the bladder, acts as a non-specific immune stimulant, inducing a potent local inflammatory response that leads to the destruction of tumor cells. However, this BCG-induced inflammation also serves as a signal for an adaptive immune resistance mechanism. Preclinical models and analyses of tumors from patients with high-risk NMIBC have demonstrated that exposure to BCG is associated with increased expression of PD-L1 on bladder cancer cells. This upregulation of PD-L1 may contribute to immune escape and subsequent BCG treatment failure by neutralizing the very T cells that BCG helps to recruit.
This understanding forms the mechanistic basis for the CREST trial. The combination of BCG and sasanlimab is designed to create a complementary, two-pronged attack. BCG acts as the "accelerator," initiating and amplifying a local anti-tumor immune response. Sasanlimab then acts to counteract the tumor's primary defense against this attack by blocking the PD-1/PD-L1 pathway, thereby preventing T-cell exhaustion and sustaining the immune-mediated assault on the cancer cells. This approach is hypothesized to yield greater and more durable anti-tumor activity than either agent could achieve alone.
Sasanlimab is a humanized IgG4-κ monoclonal antibody, engineered and produced in Chinese hamster ovary (CHO) cells. The choice of the IgG4 isotype is significant, as it is characterized by minimal Fc-dependent effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), which is intended to reduce the risk of off-target effects and improve the safety profile of the drug.
Preclinical studies have rigorously characterized the binding properties and biological activity of sasanlimab. These investigations have shown that the antibody binds selectively and with high potency to both human and cynomolgus monkey PD-1 receptors. This binding is associated with the formation of a highly stable complex, which is believed to be a primary driver of its high-affinity interaction. Critically, this binding effectively blocks the interaction of the PD-1 receptor with both of its ligands, PD-L1 and PD-L2. In vitro functional assays confirmed the biological consequences of this blockade, demonstrating that sasanlimab significantly augmented T-cell proliferation and the production of pro-inflammatory cytokines in mixed lymphocyte reaction and superantigen stimulation assays. The in vivo anti-tumor activity was confirmed in a xenogeneic model of acute graft-versus-host disease (GvHD) and, more directly, in human PD-1 knock-in mice, where sasanlimab administration halted the growth of MC-38 colon adenocarcinoma tumors.
The clinical pharmacology of sasanlimab has been evaluated in early-phase human trials. These studies have shown that the drug is active and well-tolerated in cynomolgus monkeys and demonstrates predictable pharmacokinetics in humans. A key pharmacodynamic endpoint for PD-1 inhibitors is receptor occupancy. Studies have confirmed that sasanlimab achieves full PD-1 receptor occupancy in peripheral CD8+ T cells at all clinically investigated dose levels, irrespective of the baseline PD-1 expression levels on these cells. This indicates that the chosen doses are sufficient to saturate the target and exert the intended biological effect.
The recommended clinical dose, used in the pivotal Phase III CREST trial, was established as 300 mg administered subcutaneously every 4 weeks (q4w). Further pharmacokinetic modeling and clinical study (NCT04181788) have also explored an alternative, less frequent dosing regimen of 600 mg subcutaneously every 6 weeks (q6w). This alternative regimen was found to meet the pharmacokinetic criteria for area under the curve (
AUCτ) and trough concentration (Ctrough) to support its potential use as a convenient alternative to the q4w schedule, offering greater flexibility for patients and providers.
A defining characteristic of sasanlimab that distinguishes it from many first-generation, market-leading PD-1 inhibitors is its formulation for subcutaneous (SC) administration. The drug is supplied in a prefilled syringe (2 mL) for simple injection into the adipose tissue. This contrasts with the intravenous (IV) infusion required for agents like pembrolizumab and nivolumab, which typically necessitates a visit to a specialized infusion center, insertion of an IV line, and a longer administration and observation period.
The practical implications of this difference are profound. Clinical experts and trial investigators have consistently highlighted subcutaneous delivery as a major advantage that could significantly reduce the treatment burden on both patients and the healthcare system. The simplified administration process allows for much faster patient throughput in clinics, does not require the same level of technical skill as establishing IV access, and reduces the need for dedicated infusion chairs and associated resources.
This logistical advantage may fundamentally reshape the care delivery model for NMIBC. Currently, the administration of IV immunotherapy is almost exclusively the domain of medical oncologists operating out of infusion centers. Urologists, who manage the vast majority of NMIBC care and routinely perform intravesical BCG instillations in their clinics, typically refer patients to medical oncology for systemic therapy. The simplicity of a subcutaneous injection, however, aligns seamlessly with the existing workflow of a urology practice. This could empower urologists to prescribe and administer the complete sasanlimab-BCG combination regimen themselves, creating a more integrated, "one-stop" care experience for the patient. While this presents an opportunity for more streamlined patient management, it also introduces the challenge of ensuring that urology practices are adequately equipped and trained to monitor and manage the systemic immune-related adverse events (irAEs) that can occur with PD-1 inhibitors, a responsibility that has traditionally fallen to medical oncologists. The successful implementation of this new treatment paradigm would likely require the development of new multidisciplinary care pathways and enhanced education for the urology community on irAE management.
The clinical development of sasanlimab began with a first-in-human, open-label, Phase I dose-escalation and expansion study (NCT02573259) in patients with a range of locally advanced or metastatic solid tumors. This foundational trial was designed to establish the safety, tolerability, pharmacokinetics, and preliminary anti-tumor activity of the drug. The study demonstrated that sasanlimab possessed a manageable safety profile and showed evidence of durable clinical efficacy across multiple tumor types, including malignant melanoma, squamous cell head and neck cancer, ovarian cancer, and sarcoma. These early results provided the proof-of-concept necessary to advance sasanlimab into more targeted investigations.
Following the dose-escalation phase, the Phase Ib/II portion of the study (NCT02573259) included dedicated dose expansion cohorts for patients with non-small-cell lung cancer (NSCLC) and urothelial carcinoma who were naïve to prior anti-PD-1/PD-L1 therapy. In these cohorts, patients received the recommended Phase II dose of 300 mg of sasanlimab via subcutaneous injection every 4 weeks.
These studies were instrumental not only in confirming the clinical activity of subcutaneous sasanlimab but also in exploring potential predictive biomarkers. Analyses from this trial showed that clinical response was significantly associated with higher baseline tumor mutational burden (TMB) as well as higher expression of PD-L1 and CD8. These findings are consistent with the broader class of PD-1 inhibitors and helped to validate the immunomodulatory mechanism of sasanlimab across different tumor types and routes of administration.
The clinical development program for sasanlimab extends beyond NMIBC and advanced solid tumors. Several other clinical trials are actively investigating its potential in different settings and combinations:
Non-muscle invasive bladder cancer accounts for approximately 75% of all bladder cancer diagnoses. Patients with high-risk NMIBC, defined by the presence of high-grade Ta or T1 tumors and/or carcinoma in situ (CIS), face a significant threat of disease recurrence and progression to muscle-invasive or metastatic disease. For over three decades, the global standard of care (SOC) following transurethral resection of bladder tumor (TURBT) has been intravesical immunotherapy with BCG. While many patients initially respond, disease recurrence or progression occurs in approximately 40-50% of patients within two years. For these patients, subsequent bladder-sparing treatment options are limited, and many ultimately require a radical cystectomy—the surgical removal of the bladder—which is a major operation associated with significant risks and a permanent impact on quality of life. The lack of therapeutic innovation in this frontline, BCG-naïve setting represents a major unmet clinical need.
To address this long-standing therapeutic gap, Pfizer initiated the CREST trial, a landmark study designed to evaluate the superiority of adding sasanlimab to SOC BCG. The design and key parameters of the trial are summarized in Table 1.
Table 1: Design and Arms of the Phase III CREST Trial (NCT04165317)
Parameter | Description |
---|---|
Study Title | Combination of sasanlimab and alternative BCG Regimens to Evaluate outcomes with Subcutaneous anti-PD-1 Treatment (CREST) |
Phase | Phase III |
Design | Multinational, Randomized, Open-Label, Three-Arm |
Patient Population | 1055 patients with BCG-naïve, high-risk NMIBC (High-grade Ta, T1, and/or CIS) |
Arms | Arm A (N=352): Sasanlimab 300 mg SC q4w + BCG Induction & Maintenance |
Arm B (N=352): Sasanlimab 300 mg SC q4w + BCG Induction Only | |
Arm C (N=351): BCG Induction & Maintenance (Standard of Care) | |
Primary Endpoint | Investigator-assessed Event-Free Survival (EFS) comparing Arm A vs. Arm C |
Key Secondary Endpoints | EFS (Arm B vs. Arm C), Overall Survival (OS), Complete Response (CR) Rate & Duration in CIS patients, Safety, Quality of Life (QoL) |
The trial's primary objective was to determine if the combination of sasanlimab with a full course of BCG (Arm A) could prolong EFS compared to BCG alone (Arm C). EFS was a robust composite endpoint defined as the time from randomization to the first occurrence of high-grade disease recurrence, progression of disease, persistence of CIS (for patients with CIS at baseline), or death from any cause.
The CREST trial successfully met its primary endpoint. The results, presented at major urology and oncology conferences in 2025, demonstrated that the addition of sasanlimab to standard BCG therapy led to a clinically meaningful and statistically significant improvement in EFS.
The positive primary result was supported by strong findings across key secondary endpoints and subgroups, as detailed in Table 2.
Table 2: Key Efficacy Outcomes from the CREST Trial (Arm A vs. Arm C)
Endpoint | Metric | Result |
---|---|---|
Event-Free Survival (EFS) - Primary | Hazard Ratio (95% CI); p-value | 0.68 (0.49−0.94); p=0.0095 (1-sided) |
36-Month EFS Rate | % (95% CI) | Arm A: 82.1% (77.4-85.9) vs. Arm C: 74.8% (69.7-79.2) |
EFS in T1 Subgroup | Hazard Ratio (95% CI) | 0.63 (0.41−0.96) |
EFS in CIS Subgroup | Hazard Ratio (95% CI) | 0.53 (0.29−0.98) |
CR Rate in CIS Patients (any time) | % | Arm A: 89.8% vs. Arm C: 85.2% |
Probability of Remaining in CR at 36 Months | % | Arm A: 91.7% vs. Arm C: 67.7% |
While the overall complete response rate for patients with CIS was only modestly higher in the combination arm, the durability of these responses was substantially improved. The finding that 91.7% of responders in the sasanlimab arm remained in complete response at 36 months, compared to just 67.7% in the control arm, is a particularly compelling indicator of long-term disease control. The benefit of the combination was consistent across high-risk subgroups, with even more pronounced risk reductions observed in patients with T1 disease (HR
0.63) and CIS (HR 0.53). An early interim analysis for the key secondary endpoint of overall survival (OS), conducted with a median follow-up of 40.9 months, did not show a statistically significant difference between the treatment arms; the study continues to follow patients for the final OS analysis.
The three-arm design of the CREST trial provided a crucial internal validation of the treatment strategy. The results from Arm B, which combined sasanlimab with only an induction course of BCG, showed no improvement in EFS compared to the standard of care (Arm C), which included both induction and maintenance BCG (HR 1.16; 95% CI, 0.87−1.55). This finding is highly significant. It demonstrates that the benefit of sasanlimab is not derived from simply adding it to a short course of BCG. Rather, the sustained immune stimulation provided by the full, multi-year BCG maintenance regimen is essential for sasanlimab to exert its long-term, disease-modifying effect. This underscores the biological synergy between the two agents over a prolonged period and confirms that the current standard-of-care BCG maintenance schedule should not be omitted when incorporating sasanlimab into the treatment plan.
Data from early-phase studies of sasanlimab monotherapy established a favorable and manageable safety profile. In the Phase Ib/II dose expansion cohorts involving 106 patients with NSCLC and urothelial carcinoma, grade ≥3 treatment-related adverse events (TRAEs) occurred in 13.2% of patients. The most common TRAEs of any grade included hyperthyroidism (10.4%), increased lipase (7.5%), and pruritus (7.5%). A separate Phase Ib/II study (NCT04181788) evaluating alternative dosing regimens also confirmed the drug's safety, reporting no dose-limiting toxicities (DLTs) and low rates of grade 3 TRAEs (7.3% for the 300 mg q4w dose and 3.8% for the 600 mg q6w dose), with no grade 4 or 5 TRAEs observed.
In the pivotal CREST trial, the overall safety profile of the sasanlimab-BCG combination was generally consistent with the known toxicities of each agent and the broader PD-1 inhibitor class. However, the addition of a systemic immunotherapy to local BCG therapy resulted in a notable increase in the frequency and severity of adverse events. Table 3 provides a summary of the key safety findings.
Table 3: Summary of Treatment-Related Adverse Events (TRAEs) in the CREST Trial (Arm A vs. Arm C)
Adverse Event Category | Arm A (Sasanlimab + BCG) | Arm C (BCG Alone) |
---|---|---|
Any Grade TRAEs | Higher frequency | Lower frequency |
Grade ≥3 TRAEs | ~15-17.7% | ~1.4% |
Serious TRAEs | 17.7% (62/352) | 1.4% (5/351) |
TRAEs Leading to Death | 0% | 0% |
Common Immune-Related AEs | Higher frequency, primarily endocrinopathies (e.g., thyroid dysfunction) | Low frequency |
The most significant finding was the more than tenfold increase in the rate of serious (grade ≥3) TRAEs in the combination arm (17.7%) compared to the BCG-alone arm (1.4%). Importantly, despite this increase in serious toxicity, no treatment-related deaths occurred in either arm.
As anticipated with a systemic PD-1 inhibitor, the sasanlimab-containing arms of the CREST trial saw a higher frequency of immune-related adverse events (irAEs). The most common grade 3/4 irAEs were endocrinopathies, such as hypothyroidism or hyperthyroidism, with most cases reported to be manageable and resolving with standard treatments like thyroid hormone supplementation. The increased incidence of these systemic side effects underscores the importance of careful patient monitoring and education. Expert commentary on the CREST results has consistently emphasized that successful implementation of this regimen in clinical practice will necessitate the development of robust multidisciplinary workflows to ensure the timely recognition and management of irAEs.
A critical component of evaluating a new therapy, particularly in a non-metastatic setting, is its impact on patient quality of life. Patient-reported outcome (PRO) data from the CREST trial, presented at the 2025 ASCO Annual Meeting, provided valuable context for the safety findings. Despite the objective increase in grade
≥3 adverse events, there was no clinically meaningful difference in global health scores or disease-specific symptoms (as measured by the EORTC QLQ-C30 and NMIBC24 questionnaires) between the sasanlimab-BCG arm and the BCG-alone arm.
This apparent disconnect between physician-graded toxicity and patient-reported quality of life is a crucial finding. It suggests that while the incidence of serious adverse events is higher with the combination, these events may be effectively managed without significantly detracting from the patients' overall well-being. It is plausible that the negative QoL impact of the irAEs is offset by the positive impact of improved disease control, which may lead to fewer invasive procedures (e.g., repeat biopsies, TURBTs) and reduced anxiety associated with disease recurrence. This maintenance of QoL strengthens the overall benefit-risk profile of the combination therapy and is a key consideration for patients, clinicians, and regulatory bodies.
As of September 2025, sasanlimab remains an investigational compound and has not received marketing authorization from major global health authorities, including the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA). However, the positive results from the pivotal Phase III CREST trial represent a significant milestone. Pfizer has publicly stated its intention to share these data with global regulatory authorities to support potential filings for marketing approval. If approved based on the CREST data, sasanlimab would be the first-in-class PD-1 inhibitor indicated for use in combination with BCG for the treatment of patients with BCG-naïve, high-risk NMIBC, addressing a significant unmet need and potentially establishing a new global standard of care.
The potential entry of sasanlimab into the NMIBC market requires a nuanced analysis of the competitive landscape, which includes other immunotherapies and novel intravesical agents.
The results of the CREST trial have been met with enthusiasm by key opinion leaders in the fields of urology and oncology. Dr. Neal Shore, the lead investigator for the trial, and others have described the findings as "potentially practice-changing" and a "much-needed therapeutic breakthrough," highlighting that this represents the first significant therapeutic advance in the BCG-naïve, high-risk NMIBC setting in over three decades. Experts have emphasized the clinical importance of prolonged EFS, the highly durable complete responses seen in patients with CIS, and the practical convenience afforded by the subcutaneous route of administration. At the same time, commentators have been careful to balance this optimism with a pragmatic view of the increased toxicity. The higher rate of serious and immune-related adverse events is acknowledged as a critical factor that will require careful patient selection and shared decision-making, as well as the establishment of multidisciplinary care teams to effectively manage potential side effects.
Despite the success of the CREST trial, several important questions remain that will guide future research. The most prominent is the lack of a demonstrated overall survival benefit in the interim analysis. While EFS is a well-accepted endpoint in NMIBC, longer-term follow-up from the trial will be essential to determine if the observed delay in recurrence ultimately translates into improved survival.
Another critical area for future investigation is the identification of predictive biomarkers. The subgroup analyses from CREST revealed that baseline PD-L1 expression was not predictive of which patients benefited from the addition of sasanlimab. This finding, while disappointing, is not uncommon for checkpoint inhibitors in bladder cancer and highlights the urgent need for more sophisticated biomarkers—potentially involving gene expression signatures, TMB, or ctDNA analysis—to better select patients who are most likely to respond and to spare those who are unlikely to benefit from the added toxicity and cost of systemic immunotherapy.
Sasanlimab represents a significant advancement in the field of immuno-oncology, with a robust clinical development program culminating in the positive results of the pivotal Phase III CREST trial. The combination of subcutaneous sasanlimab with standard-of-care intravesical BCG has demonstrated a statistically significant and clinically meaningful reduction in the risk of disease recurrence or progression for patients with high-risk, BCG-naïve non-muscle invasive bladder cancer. The therapy's compelling efficacy, particularly the remarkable durability of complete responses in patients with carcinoma in situ, is complemented by a safety profile that, while more toxic than BCG alone, is considered manageable within the context of its clinical benefit and is not associated with a detriment to overall quality of life.
The strategic advantages of its subcutaneous administration and its positioning in an earlier line of therapy create a distinct and valuable role for sasanlimab in the evolving NMIBC treatment landscape. Pending regulatory approval, the sasanlimab-BCG combination is poised to establish a new global standard of care, offering a vital new bladder-sparing option for patients and marking the first major therapeutic innovation in this setting in over 30 years. Future research will be critical to confirm a long-term survival benefit and to identify predictive biomarkers to optimize patient selection and further refine its role in personalized cancer care.
Published at: September 25, 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.