Small Molecule
C24H28N6O3S
1308672-74-3
Surufatinib is a novel, orally administered small-molecule drug developed by HUTCHMED for the treatment of solid tumors. It is distinguished by a unique dual mechanism of action as an "angio-immuno" kinase inhibitor, concurrently targeting pathways involved in tumor angiogenesis and the immune-suppressive tumor microenvironment. Specifically, it potently inhibits vascular endothelial growth factor receptors (VEGFR) 1, 2, and 3, and fibroblast growth factor receptor 1 (FGFR1) to block the formation of new blood vessels, while also inhibiting colony-stimulating factor-1 receptor (CSF-1R) to modulate the activity of tumor-promoting macrophages.
The clinical value of surufatinib has been robustly demonstrated in two pivotal, randomized, placebo-controlled Phase III trials conducted in China. The SANET-ep trial showed that surufatinib significantly improved median progression-free survival (PFS) to 9.2 months versus 3.8 months for placebo in patients with advanced extra-pancreatic neuroendocrine tumors (epNETs). Similarly, the SANET-p trial demonstrated a median PFS of 10.9 months versus 3.7 months for placebo in patients with advanced pancreatic neuroendocrine tumors (pNETs). These compelling results, which led to both trials being stopped early for efficacy, formed the basis for surufatinib's approval in China under the brand name Sulanda®, where it is now an important treatment option for patients with NETs.
In stark contrast to its success in China, surufatinib has faced significant regulatory setbacks in Western markets. In 2022, the U.S. Food and Drug Administration (FDA) issued a Complete Response Letter (CRL), declining to approve the drug based on a data package comprising the two Chinese trials and a small U.S. bridging study. The FDA cited the need for a multi-regional clinical trial (MRCT) that is more representative of the U.S. patient population and medical practice. Shortly thereafter, HUTCHMED withdrew its Marketing Authorization Application (MAA) from the European Medicines Agency (EMA) after receiving feedback that raised similar concerns about the applicability of the single-country data and compliance with Good Clinical Practice.
The story of surufatinib is therefore one of a drug with proven clinical efficacy but a challenging global regulatory journey. It serves as a critical case study in the evolving landscape of global drug development, highlighting a paradigm shift where data generated exclusively in China, even when scientifically robust, is no longer considered sufficient for approval in the U.S. and Europe without a comprehensive MRCT. The future of surufatinib outside of China is contingent on HUTCHMED's ability to navigate these heightened regulatory expectations, while its development continues in combination therapies for other aggressive cancers.
This section establishes the fundamental identity of surufatinib, providing the necessary chemical, physical, and commercial context for the compound.
Surufatinib (DrugBank ID: DB15106) is a small molecule inhibitor of multiple receptor tyrosine kinases [User Query]. The compound was previously known in literature and early development as sulfatinib and is also identified by its development code, HMPL-012.[1] Its Chemical Abstracts Service (CAS) Registry Number is 1308672-74-3.[3] This consistent nomenclature is critical for accurately tracking the drug's development and research history across various scientific and regulatory documents.
Surufatinib's chemical identity is well-defined. Its formal International Union of Pure and Applied Chemistry (IUPAC) name is N-(2-(dimethylamino)ethyl)-1-(3-((4-((2-methyl-1H-indol-5-yl)oxy)pyrimidin-2-yl)amino)phenyl)methanesulfonamide.[3] Physically, it is described as an off-white solid.[3] The molecular formula is
C24H28N6O3S, corresponding to a molecular weight of 480.59 g/mol.[3] The consistency of these foundational chemical data points across multiple independent chemical suppliers and scientific databases provides a high degree of confidence in the compound's identity, which is the bedrock upon which all reliable preclinical and clinical investigations are built.[3]
For clinical use, surufatinib is administered orally in the form of capsules.[6] Its solubility profile is characteristic of many small molecule kinase inhibitors, showing good solubility in organic solvents like dimethyl sulfoxide (DMSO) at 10 mg/ml and dimethylformamide (DMF) at 30 mg/ml, but poor solubility in ethanol (1 mg/ml) and aqueous solutions like phosphate-buffered saline (PBS) at pH 7.2 (0.12 mg/ml).[4] This solubility profile dictates the requirements for its formulation to ensure adequate oral bioavailability. The compound demonstrates good chemical stability, with a shelf life of at least four years when stored at -20°C.[4]
Surufatinib was discovered and developed by HUTCHMED (China) Limited, formerly known as Hutchison China MediTech (Chi-Med).[6] HUTCHMED retains all worldwide rights to the drug.[9] In China, where it has received regulatory approval, surufatinib is marketed under the commercial brand name
Sulanda®.[5] For its European regulatory submission, the proposed trade name was
Sevsury, though this application was ultimately withdrawn.[13]
Table 1: Chemical and Physical Properties of Surufatinib
Property | Value | Source(s) |
---|---|---|
Common Name | Surufatinib | [User Query] |
Synonyms | Sulfatinib, HMPL-012 | 1 |
DrugBank ID | DB15106 | [User Query] |
CAS Number | 1308672-74-3 | 1 |
Type | Small Molecule | [User Query] |
IUPAC Name | N-(2-(dimethylamino)ethyl)-1-(3-((4-((2-methyl-1H-indol-5-yl)oxy)pyrimidin-2-yl)amino)phenyl)methanesulfonamide | 3 |
Molecular Formula | C24H28N6O3S | 3 |
Molecular Weight | 480.59 g/mol | 3 |
Developer | HUTCHMED (China) Limited | 6 |
Brand Name (China) | Sulanda® | 5 |
Proposed Brand Name (EU) | Sevsury | 13 |
Surufatinib is characterized by a unique and sophisticated mechanism of action that targets two distinct but complementary pillars of cancer progression: tumor angiogenesis and immune evasion. This has led to its classification as a novel "angio-immuno" kinase inhibitor.[2]
The "angio" component of surufatinib's activity stems from its potent inhibition of key receptor tyrosine kinases that drive angiogenesis—the process by which tumors form new blood vessels to secure oxygen and nutrients for growth and metastasis.[2] Surufatinib targets:
The potency of this inhibition is demonstrated by its low half-maximal inhibitory concentrations (IC50), with values of 2 nM for VEGFR1, 24 nM for VEGFR2, 1 nM for VEGFR3, and 15 nM for FGFR1.[4] By blocking these pathways, surufatinib effectively aims to cut off the tumor's blood supply, thereby inhibiting its growth and spread.[6]
The "immuno" component of surufatinib's mechanism targets the tumor's ability to evade the host immune system. This is achieved through the inhibition of:
The CSF-1/CSF-1R signaling axis is critical for the recruitment, differentiation, and survival of macrophages.[2] Within the tumor microenvironment, this pathway promotes the accumulation of tumor-associated macrophages (TAMs), a population of immune cells that are typically polarized to support tumor growth, promote angiogenesis, facilitate metastasis, and suppress the anti-tumor activity of other immune cells, such as T-cells.[2] By inhibiting CSF-1R, surufatinib is designed to reduce the population of these pro-tumorigenic TAMs, thereby remodeling the tumor microenvironment to be less immunosuppressive and potentially more susceptible to immune-mediated destruction.[2]
This dual mechanism is strategically synergistic. It is well-established that resistance to anti-angiogenic therapies can arise through tumor adaptation, where hypoxia induced by VEGFR inhibition can trigger the recruitment of pro-angiogenic immune cells, including TAMs, which then restore blood vessel growth.[2] By simultaneously targeting both VEGFR/FGFR1 and CSF-1R, surufatinib delivers a "one-two punch": it directly attacks the tumor's vascular supply while also preemptively blocking a key immune-mediated escape mechanism. This sophisticated design provides a strong rationale for its potential synergy with other immunotherapies like PD-1 checkpoint inhibitors, as it may help create a more "inflamed" tumor microenvironment that is primed for an effective anti-tumor immune response.[15]
Furthermore, surufatinib exhibits high selectivity. In a broad kinase screen, it was found to be selective for its intended targets over a panel of 278 other kinases, suggesting a focused mechanism with a lower potential for off-target toxicities.[4]
The pharmacokinetic (PK) profile of surufatinib has been characterized in both preclinical models and human studies, supporting its clinical dosing regimen.
Pharmacodynamic studies have confirmed that surufatinib engages its intended targets in patients. Treatment has been shown to cause measurable changes in circulating biomarkers consistent with its mechanism of action, including increases in plasma VEGF-A and FGF23, and decreases in soluble VEGFR-2 (sVEGFR-2).[2] These biomarker modulations serve as in-vivo proof of target engagement and confirm that the drug is exerting its expected biological effects.
Table 2: Key Pharmacokinetic Parameters of Surufatinib in Humans
PK Parameter | Value | Source(s) |
---|---|---|
Route of Administration | Oral | 6 |
Recommended Dose | 300 mg once daily (QD) | 2 |
Time to Max Concentration (Tmax) | ~4 hours | 18 |
Elimination Half-life (t1/2) | ~23.3 hours | 18 |
Primary Route of Excretion | Fecal (~87%) | 18 |
Major Circulating Moiety | Parent Drug | 18 |
The clinical efficacy of surufatinib in its primary indication of advanced neuroendocrine tumors (NETs) is defined by two large, pivotal Phase III trials conducted in China—SANET-ep and SANET-p—and supported by a U.S. bridging study. The outcomes of these trials established surufatinib as a highly active agent in this disease.
The SANET-ep study was a randomized, double-blind, placebo-controlled Phase III trial designed to evaluate the efficacy and safety of surufatinib in patients with advanced, progressive, well-differentiated (Grade 1 or 2) extra-pancreatic neuroendocrine tumors (epNETs).[20] The trial enrolled 198 patients in China who were randomized in a 2:1 ratio to receive either oral surufatinib 300 mg once daily (n=129) or a matching placebo (n=69).[20]
In a testament to the drug's significant activity, the trial was stopped early in June 2019 at a pre-planned interim analysis after the Independent Data Monitoring Committee (IDMC) concluded that it had met its primary endpoint with overwhelming efficacy.[20]
Running in parallel to SANET-ep, the SANET-p study was a similarly designed randomized, double-blind, placebo-controlled Phase III trial focused on patients with advanced, progressive, well-differentiated pancreatic neuroendocrine tumors (pNETs).[25] The trial randomized 172 patients in China (2:1) to receive surufatinib 300 mg QD (n=113) or placebo (n=59).[25]
Like its counterpart, the SANET-p trial was also stopped early (in January 2020) on the recommendation of its IDMC due to compelling evidence of efficacy at a planned interim analysis.[25]
To support a regulatory submission in the United States, HUTCHMED conducted a dose-escalation and expansion study in U.S. patients. The primary goal was to "bridge" the data from the Chinese SANET trials by confirming the safety, PK, and preliminary efficacy of the 300 mg QD dose in a Western population.[19]
The study enrolled a cohort of 32 patients with heavily pre-treated, progressive NETs (16 pNET, 16 epNET). This was a challenging population to treat, with a median of three prior lines of therapy, and all patients had previously received the standard-of-care targeted agents everolimus and/or sunitinib.[19]
Despite the advanced nature of the disease in this cohort, surufatinib demonstrated promising anti-tumor activity:
The robust efficacy demonstrated in the large, randomized SANET trials is undeniable. Both studies met the highest standard of clinical evidence, with IDMCs independently recommending early termination due to the clear benefit of surufatinib over placebo. The magnitude of PFS improvement—5.4 months in epNETs and 7.2 months in pNETs—is highly clinically meaningful for patients with advanced cancer. Furthermore, the U.S. bridging study, though small, suggested that the drug was at least as effective, and perhaps even more so, in a Western patient population that had already exhausted standard therapies. This strong and consistent evidence base across three distinct trials created a significant point of tension and surprise when Western regulatory agencies later rejected the drug, shifting the narrative from one of clinical success to one of regulatory strategy.
Table 3: Pivotal Phase III Trial Efficacy Results (SANET-p & SANET-ep)
Endpoint | SANET-ep (Extra-Pancreatic NETs) | SANET-p (Pancreatic NETs) | Source(s) |
---|---|---|---|
Surufatinib (n=129) | Placebo (n=69) | Surufatinib (n=113) | |
Median PFS (months) | 9.2 | 3.8 | 10.9 |
Hazard Ratio (95% CI) | 0.334 (0.223–0.499) | 0.491 (0.32–0.76) | 22 |
p-value | <0.0001 | 0.0011 | 22 |
Objective Response Rate (ORR) | 10.3% | 0% | 19.2% |
Disease Control Rate (DCR) | 86.5% | 65.7% | 80.8% |
The safety profile of surufatinib has been extensively characterized through its clinical trial program, with the most robust data derived from a pooled analysis of the two pivotal SANET trials. The observed adverse events are largely consistent with the known class effects of potent VEGFR inhibitors and were deemed manageable.
A pooled analysis of the SANET-ep and SANET-p trials, encompassing 263 patients treated with surufatinib and 133 with placebo, provides a comprehensive overview of the drug's safety.[29]
The safety profile is a direct reflection of the drug's mechanism of action. Hypertension and proteinuria are well-established class effects associated with potent inhibition of the VEGFR pathway. Their high incidence with surufatinib is therefore not unexpected and falls within the spectrum of toxicities oncologists are accustomed to managing with this class of drugs. This predictability was a key factor in the assessment that the drug's risk-benefit profile was favorable, an assessment underscored by regulatory bodies, which explicitly stated that their non-approval decisions were not related to safety issues.[30]
Table 4: Common and Grade ≥3 Adverse Events from Pooled SANET Analysis
Adverse Event | Surufatinib (Any Grade %) | Placebo (Any Grade %) | Surufatinib (Grade ≥3 %) | Placebo (Grade ≥3 %) |
---|---|---|---|---|
Proteinuria | 68.8 | 54.9 | 14.8 | 0.8 |
Hypertension | 68.4 | 27.1 | 38.8 | 13.5 |
Diarrhea | 49.0 | 22.6 | 2.3 | 0.8 |
Blood TSH Increased | 42.6 | 9.8 | 0 | 0 |
Blood Bilirubin Increased | 37.6 | 19.5 | 1.9 | 0 |
Hypertriglyceridemia | 35.7 | 12.0 | 5.3 | 0 |
Source: 29 |
Although adverse events were frequent, they were generally manageable with proactive monitoring and intervention. The median time to onset for both hypertension and proteinuria was less than one month, highlighting the need for early and regular monitoring of blood pressure and urinalysis upon treatment initiation.[29]
Dose modification was a key strategy for managing toxicity. In the pooled analysis, TEAEs led to dose reductions in 43.0% of patients and dose interruptions in 47.1%.[29] A real-world study similarly found that approximately one-third of patients required a dose reduction from the standard 300 mg daily dose to 250 mg or 200 mg, which in most cases successfully mitigated the adverse events.[23] Despite the high frequency of dose modifications, the majority of patients (83.3%) were able to continue treatment without permanent discontinuation due to AEs, indicating that these toxicities, while common, did not typically necessitate stopping this effective therapy.[29]
Based on its mechanism of action and observed toxicities, surufatinib has several contraindications and potential drug-drug interactions (DDIs).
The regulatory history of surufatinib is a tale of two vastly different outcomes, marked by decisive success in China and comprehensive rejection in the West. This divergence provides a critical lens through which to view the current landscape of global pharmaceutical development and regulation.
Leveraging the compelling, positive results from the domestically conducted SANET-ep and SANET-p trials, HUTCHMED achieved regulatory success in China. The National Medical Products Administration (NMPA) granted two separate approvals for surufatinib, marketed as Sulanda®:
These approvals represented a major corporate milestone for HUTCHMED, as surufatinib became its first self-discovered oncology drug to reach the market without a development partner.[11] The drug was subsequently included in China's National Reimbursement Drug List (NRDL) in January 2022, ensuring broad patient access within the country.[12]
HUTCHMED's strategy for U.S. approval was based on an agreement reached with the FDA in a pre-New Drug Application (NDA) meeting in May 2020. The understanding was that the two positive Chinese Phase III studies, supported by data from the U.S. bridging study, could form the basis of an NDA submission.[31] The NDA was subsequently submitted and accepted for review by the FDA in July 2021, with a target action date of April 30, 2022.[15]
However, on May 2, 2022, HUTCHMED announced it had received a Complete Response Letter (CRL) from the FDA, formally rejecting the application in its current form.[30] The reasons cited by the FDA were not related to the drug's safety or efficacy per se, but rather the nature and origin of the evidence package:
A similar story unfolded in Europe. HUTCHMED's Marketing Authorization Application (MAA) for surufatinib was validated by the EMA in July 2021.[8] However, on August 1, 2022, the company formally withdrew the application before a final decision was rendered.[13]
The withdrawal was prompted by feedback from the EMA's Committee for Medicinal Products for Human Use (CHMP), which had communicated a provisional opinion that the drug could not be authorized. The concerns raised by the EMA mirrored those of the FDA and were even more detailed:
In its withdrawal letter, HUTCHMED acknowledged that it was unable to satisfactorily address these fundamental concerns within the procedural timelines.[13]
The regulatory rejections of surufatinib in both the U.S. and Europe, despite the existence of a prior agreement with the FDA on the submission package, represent a significant paradigm shift. This case, along with the similar rejection of the PD-1 inhibitor sintilimab, serves as a landmark event signaling a clear tightening of evidentiary standards by Western regulators for drugs developed primarily or exclusively in China. While earlier drugs had gained approval based on China-only data (e.g., zanubrutinib), the bar has now been raised significantly.[40] The surufatinib story is a cautionary tale for the global pharmaceutical industry, demonstrating that the strategy of using single-country data supplemented by a small bridging study is no longer a viable path to approval in the U.S. or EU for most indications. The clear message is that future drug development intended for global markets must incorporate MRCTs from an early stage, a far more complex and costly endeavor.
Table 5: Global Regulatory Submission and Outcome Summary
Regulatory Agency | Action | Date | Outcome / Reason | Source(s) |
---|---|---|---|---|
NMPA (China) | epNET Approval | Dec 2020 | Approved based on SANET-ep trial results. | 7 |
pNET Approval | Jun 2021 | Approved based on SANET-p trial results. | 9 | |
FDA (U.S.) | NDA Accepted | Jul 2021 | PDUFA date set for April 30, 2022. | 15 |
Complete Response Letter | May 2022 | Rejected. Required a multi-regional clinical trial (MRCT); cited concerns over applicability of single-country data to U.S. population and medical practice. | 30 | |
EMA (Europe) | MAA Validated | Jul 2021 | Formal review commenced. | 8 |
MAA Withdrawn | Aug 2022 | Withdrawn by applicant. EMA had concerns about study design, GCP compliance, and manufacturing. Provisional opinion was that benefits did not outweigh risks. | 13 |
To understand the clinical potential of surufatinib, it is essential to position it within the existing therapeutic landscape for advanced NETs by comparing its efficacy against established treatments.
A retrospective, observational study conducted at the Fudan University Shanghai Cancer Center provides the most direct, albeit non-randomized, comparison of surufatinib against the standard-of-care targeted therapies sunitinib and everolimus in a real-world setting.[41]
While direct comparisons from randomized trials are lacking, a cross-trial comparison of pivotal study data provides valuable context.
The collective data suggests that surufatinib is a highly competitive agent in the treatment of progressive, well-differentiated NETs. Its median PFS of 10.9 months in pNETs from the SANET-p trial is directly comparable to the 11.0-11.4 month PFS demonstrated by everolimus and sunitinib in their respective pivotal trials. The real-world data from China further strengthens this position, suggesting potential superiority.[41] The comparison with lanreotide is less direct, as lanreotide is typically used in patients with less aggressive, slower-growing tumors, leading to longer observed PFS.
The critical question, as noted by experts, is where surufatinib fits into the complex treatment algorithm for NETs.[38] The data from the U.S. bridging study, which showed efficacy in patients who had already failed everolimus and/or sunitinib, suggests that surufatinib has activity in a later-line setting.[28] Its unique CSF-1R inhibitory activity might also position it as a preferred agent for certain patient subgroups or as an ideal partner for combination with immunotherapy.
However, without regulatory approval in the U.S. or Europe, surufatinib's role in the global treatment algorithm remains purely theoretical. Its use is confined to China and ongoing clinical trials, leaving a significant unmet need for additional treatment options for NET patients in Western countries.
Despite its regulatory setbacks in the West for neuroendocrine tumors, the development of surufatinib continues, with a strategic focus on leveraging its unique angio-immuno mechanism in other malignancies and in combination therapies.
HUTCHMED is actively pursuing the development of surufatinib in several other cancer types, primarily as part of combination regimens designed to exploit potential synergies.
This development strategy underscores a clear shift towards combination therapies, where surufatinib's ability to remodel the tumor microenvironment may enhance the efficacy of both immunotherapy and chemotherapy.
For the NET indication in the U.S. and Europe, the path forward is narrow and challenging. The FDA has explicitly stated that a new Multi-Regional Clinical Trial (MRCT) is required for approval.[31] Such a trial would need to be designed from the outset to include a diverse, global patient population representative of Western countries. Furthermore, given the availability of effective treatments like everolimus and sunitinib, it would likely need to use an active comparator as its control arm rather than placebo. This represents a substantial financial and operational investment for HUTCHMED, and the company's decision on whether to undertake such a trial will ultimately determine the drug's fate in Western markets.[40]
Surufatinib is a well-characterized small-molecule inhibitor with a rational and sophisticated dual mechanism of action targeting both tumor angiogenesis and immune evasion. Its clinical efficacy in advanced, well-differentiated neuroendocrine tumors is supported by high-quality evidence from two large, randomized, placebo-controlled Phase III trials, which demonstrated clinically meaningful improvements in progression-free survival with a predictable and manageable safety profile.
However, the story of surufatinib serves as a powerful and cautionary case study in the complexities of modern global drug development. Its regulatory failures in the United States and Europe were not a reflection of the drug's intrinsic clinical value, but rather a consequence of an evidence package that, while sufficient for approval in China, was deemed inadequate by Western agencies. These agencies have demonstrated a clear and heightened expectation for multi-regional clinical trial data that is directly applicable to their diverse patient populations and standards of care.
Consequently, the future of surufatinib is bifurcated. In China, it is an established and valuable therapeutic option for patients with NETs. Globally, its potential in this indication remains unrealized, contingent upon the successful completion of a new and costly MRCT. The most promising path forward for surufatinib on the international stage may lie in its ongoing development in combination therapies for other aggressive cancers, provided these new, ambitious trials are designed from their inception to meet the rigorous evidentiary standards of a global regulatory environment.
Published at: August 7, 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.