scholarly journals Phase II Study of Thalidomide Plus Dexamethasone Induction Followed by Tandem Melphalan-Based Autotransplantation and Thalidomide-Plus-Prednisone Maintenance for Untreated Multiple Myeloma: A Southwest Oncology Group Trial (S0204)

2009 ◽  
Vol 27 (21) ◽  
pp. 3510-3517 ◽  
Author(s):  
Mohamad A. Hussein ◽  
Vanessa Bolejack ◽  
Jeffrey A. Zonder ◽  
Brian G.M. Durie ◽  
Andrzej J. Jakubowiak ◽  
...  

Purpose Thalidomide-dexamethasone (THAL-DEX) is standard induction therapy for multiple myeloma (MM). Tandem melphalan-based transplantations have yielded superior results to single transplantations. Phase II trial S0204 was designed to improve survival results reported for the predecessor, phase III trial S9321 by 50%. Patients and Methods Newly diagnosed patients with MM were eligible for S0204 with THAL-DEX induction, tandem melphalan-based tandem transplantation, and THAL-prednisone maintenance. Results Of 143 eligible patients, 142 started induction, 73% completed first transplantation, 58% completed second transplantation, and 56% started maintenance. The quantity of stem cells required for two transplantations was reached in 88% of 111 patients undergoing collection, 74% of whom completed both transplantations. Partial response, very good partial remission, and complete response were documented after 12 months of maintenance therapy in 87%, 72%, and 22% of patients, respectively. During a median follow-up time of 37 months, 4-year estimates of event-free and overall survival were 50% and 64%, respectively. Survival outcomes were superior for International Staging System (ISS) stage 1 disease, when lactate dehydrogenase (LDH) levels were normal and a second transplantation was applied in a timely fashion. Conclusion Both overall survival (P = .0002) and event-free survival (P < .0001) were significantly improved with S0204 compared with S9321 when 121 and 363 patients, respectively, were matched on ISS stage and LDH.

2021 ◽  
pp. JCO.21.00972
Author(s):  
Paul G. Richardson ◽  
Shaji K. Kumar ◽  
Tamás Masszi ◽  
Norbert Grzasko ◽  
Nizar J. Bahlis ◽  
...  

PURPOSE The double-blind, placebo-controlled, phase III TOURMALINE-MM1 study demonstrated a statistically significant improvement in progression-free survival with ixazomib-lenalidomide-dexamethasone (ixazomib-Rd) versus placebo-Rd in patients with relapsed or refractory multiple myeloma. We report the final analyses for overall survival (OS). PATIENTS AND METHODS Patients were randomly assigned to ixazomib-Rd (n = 360) or placebo-Rd (n = 362), stratified by number of prior therapies (1 v 2 or 3), previous proteasome inhibitor (PI) exposure (yes v no), and International Staging System disease stage (I or II v III). OS (intent-to-treat population) was a key secondary end point. RESULTS With a median follow-up of 85 months, median OS with ixazomib-Rd versus placebo-Rd was 53.6 versus 51.6 months (hazard ratio, 0.939; P = .495). Lower hazard ratios, indicating larger magnitude of OS benefit with ixazomib-Rd versus placebo-Rd, were seen in predefined subgroups: refractory to any (0.794) or last (0.742) treatment line; age > 65-75 years (0.757); International Staging System stage III (0.779); 2/3 prior therapies (0.845); high-risk cytogenetics (0.870); and high-risk cytogenetics and/or 1q21 amplification (0.862). Following ixazomib-Rd versus placebo-Rd, 71.7% versus 69.9% of patients received ≥ 1 anticancer therapy, of whom 24.7% versus 33.9% received daratumumab and 71.8% versus 76.9% received PIs (next-line therapy: 47.5% v 55.8%). Rates of new primary malignancies were similar with ixazomib-Rd (10.3%) and placebo-Rd (11.9%). There were no new or additional safety concerns. CONCLUSION Median OS values in both arms were the longest reported in phase III studies of Rd-based triplets in relapsed or refractory multiple myeloma at the time of this analysis; progression-free survival benefit with ixazomib-Rd versus placebo-Rd did not translate into a statistically significant OS benefit on intent-to-treat analysis. OS benefit was greater in subgroups with adverse prognostic factors. OS interpretation was confounded by imbalances in subsequent therapies received, especially PIs and daratumumab.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5167-5167
Author(s):  
Shingo Kurahashi ◽  
Hiroto Narimatsu ◽  
Takumi Sugimoto ◽  
Isamu Sugiura

Abstract Introduction: Since multiple myeloma (MM) is not a curative disease and clinical outcome is variable, chemotherapy is started only when patients developed organ impairment or progression of disease. As for clinical staging, Durie&Salmon (DS) system is in use. The International Staging System (ISS) for MM has been recently reported to provide simple and useful prognostic grouping (Greipp et al. 2005). However, its usefulness in therapeutic strategy has not been clearly demonstrated. Patients and methods: We reviewed medical records of patients with MM, newly diagnosed in Toyohashi Municipal Hospital between May 1997 and April 2004. They were all stratified based on both ISS and DS system. Results: The median age of 55 patients was 67 years (range; 46–86). M protein isotypes included IgG (n=33), IgA (n=13), BJP (n=6) and IgD (n=1). Fifty-two patients were treated with chemotherapy and 12 of those patients underwent autologous peripheral blood stem cell transplantation. The median follow-up of the patients was 26.8 months (range; 1.4–77.5). Their staging and overall survival (OS) are summarized on the following table. ISS predicted OS more clearly than DS system in our study. Overall survival based on ISS and DS system ISS DS stage no. of patients OS at 3 yrs no. of patients OS at 3 yrs I 14 1.00 3 0.67 II 22 0.55 20 0.79 III 19 0.25 32 0.40 p-value 0.0102 0.3287 Thirteen of the 14 patients with ISS stage I are alive at median months of 31.5 (10.4–73.8), and only one patient died of secondary malignancy at 44.9 months from diagnosis. The patients with ISS stage I included 86% of DS stage II and III patients, who are usually required treatment. Conclusions and discussions: ISS could predict clinical outcome more clearly than DS system. The patients’ prognosis was good in ISS stage I although many patients with DS stage II and III was included in this group. We suggests that early treatment to the patients with ISS stage I might not be necessarily required. Further studies are needed to adopt ISS instead of DS system in therapeutic strategy. Figure Figure


2014 ◽  
Vol 32 (25) ◽  
pp. 2712-2717 ◽  
Author(s):  
Murielle Roussel ◽  
Valérie Lauwers-Cances ◽  
Nelly Robillard ◽  
Cyrille Hulin ◽  
Xavier Leleu ◽  
...  

Purpose The three-drug combination of lenalidomide, bortezomib, and dexamethasone (RVD) has shown significant efficacy in multiple myeloma (MM). The Intergroupe Francophone du Myélome (IFM) decided to evaluate RVD induction and consolidation therapies in a sequential intensive strategy for previously untreated transplantation-eligible patients with MM. Patients and Methods In this phase II study, 31 symptomatic patients age < 65 years were enrolled to receive three RVD induction cycles followed by cyclophosphamide harvest and transplantation. Patients subsequently received two RVD consolidation cycles and 1-year lenalidomide maintenance. Results Very good partial response rate or better at the completion of induction, transplantation, and consolidation therapy was 58%, 70%, and 87%, respectively. Maintenance upgraded responses in 27% of patients. Overall, 58% of patients achieved complete response, and 68% were minimal residual disease (MRD) negative by flow cytometry. The most common toxicities with RVD were neurologic and hematologic, including grade 1 to 2 sensory neuropathy (55%), grade 3 to 4 neutropenia (35%), and thrombocytopenia (13%). Two basal cell carcinomas in the same patient and one case of breast cancer were observed. There was no treatment-related mortality. With a median follow-up of 39 months, estimated 3-year progression-free and overall survival were 77% and 100%, respectively. None of the patients who achieved MRD negativity relapsed. Conclusion The transplantation program with RVD induction and consolidation followed by lenalidomide maintenance produced high-quality responses and showed favorable tolerability in patients with newly diagnosed MM. Overall, 68% of patients achieved MRD negativity; none of these patients relapsed. This program is being evaluated in the ongoing IFM/Dana-Farber Cancer Institute 2009 phase III study.


2014 ◽  
Vol 32 (29) ◽  
pp. 3299-3306 ◽  
Author(s):  
Sant P. Chawla ◽  
Lee D. Cranmer ◽  
Brian A. Van Tine ◽  
Damon R. Reed ◽  
Scott H. Okuno ◽  
...  

Purpose TH-302, a prodrug of the cytotoxic alkylating agent bromo-isophosphoramide mustard, is preferentially activated in hypoxic conditions. This phase II study investigated TH-302 in combination with doxorubicin, followed by single-agent TH-302 maintenance therapy in patients with first-line advanced soft tissue sarcoma (STS) to assess progression-free survival (PFS), response rate, overall survival, safety, and tolerability. Patients and Methods In this open-label phase II study, TH-302 300 mg/m2 was administered intravenously on days 1 and 8 with doxorubicin 75 mg/m2 on day 1 of each 21-day cycle. After six cycles, patients with stable and/or responding disease could receive maintenance monotherapy with TH-302. Results Ninety-one patients initiated TH-302 plus doxorubicin induction treatment. The PFS rate at 6 months (primary efficacy measure) was 58% (95% CI, 46% to 68%). Median PFS was 6.5 months (95% CI, 5.8 to 7.7 months); median overall survival was 21.5 months (95% CI, 16.0 to 26.2 months). Best tumor responses were complete response (n = 2 [2%]) and partial response (n = 30 [34%]). During TH-302 maintenance (n = 48), five patients improved from stable disease to partial response, and one patient improved from partial to complete response. The most common adverse events during induction were fatigue, nausea, and skin and/or mucosal toxicities as well as anemia, thrombocytopenia, and neutropenia. These were less severe and less frequent during maintenance. There was no evidence of TH-302–related hepatic, renal, or cardiac toxicity. Conclusion PFS, overall survival, and tumor response compared favorably with historical outcomes achieved with other first-line chemotherapies for advanced STS. A phase III study of TH-302 is ongoing (NCT01440088).


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1635-1635
Author(s):  
Yuxin Liu ◽  
Bohm Kywe ◽  
Lake Crawford ◽  
Federico Lora ◽  
Noffar Bar ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is an incurable yet highly heterogeneous disease of clonal plasma cells. The role of induction therapy is to achieve a complete response, thereby reducing the number of malignant plasma cells in the bone marrow. Partial responses or less are considered suboptimal and are associated with poorer outcomes. Following the approval of monoclonal antibody (mAb) therapies in MM, little is known of their effect on primary refractory and suboptimal responses in real-world populations. Methods: This retrospective cohort study included patients with clinical MM who were diagnosed and treated within the Yale New Haven Health System between 2009-2019. Our primary aim was to evaluate the overall survival (OS) in patients with primary refractory (PriRef) or suboptimal responding disease compared to those who achieved at least a very good partial response (≥VGPR). PriRef disease was assessed following three months of induction therapy, and was defined as a partial response, stable disease, or progressive disease based on International Myeloma Working Group (IMWG) criteria. A stricter cutoff of less than a VGPR was chosen for our definition of PriRef, because deeper responses, including MRD-negative status, have demonstrated increasingly important prognostic value. Descriptive statistics and OS analysis were performed with 2-tailed contingency χ²-tests, t-tests, Kaplan Meier, and log-rank tests using GraphPad Prism v8. Results: 246 patients (pts) with adequate response data following 3 months of induction therapy were identified. Clinical features included median age at diagnosis of 61 years (range, 34-92), 26.8% of whom were 70 years or older, 54.9% male, 63.4% Caucasian, 24.3% Black or African American, and 11% Hispanic. 46.8% had International Staging System (ISS) stage II or III disease and 41.1% had Revised International Staging System (R-ISS) stage II or III disease. 33.3% of patients had high-risk cytogenetics, which was defined as a del17/17p, t(4:14), t(4;16), t(4;20), gain or amplification of 1q, deletion of 1p, and chromosome 1 translocations. 81.7% of this cohort received a triplet or quadruplet induction regimen. The majority of patients received triplet induction regimens that included immunomodulatory and proteasome inhibitor combinations, and only two patients were prescribed quadruplet therapy. 44.7% of patients received autologous stem cell transplant (ASCT) at some point in their treatment course, with 79.1% (87/110) in the upfront setting and 20.9% (23/110) having delayed ASCT (Table 1). In this cohort, 122 pts (49.6%) achieved ≥VGPR and 124 pts (50.4%) were PriRef. There were no significant differences in the baseline demographics, ISS, R-ISS, or cytogenetic characteristics between ≥VGPR and PriRef. PriRef patients were less likely to have been treated with at least a triplet induction regimen than ≥VGPR (74.2% vs. 89.3%, p &lt;0.01), and they were also less likely to have undergone ASCT (37.1% % vs. 52.5%, p = 0.02). When the two cohorts were stratified by upfront versus delayed ASCT, PriRef pts still demonstrated lower rates of ASCT in both settings, but the differences were not statistically significant (Table 1). There was no significant difference in OS between PriRef and ≥VGPR pts. Median OS for PriRef was 102.9 months compared to 114.9 months in ≥VGPR (p = 0.27) (Figure 1). Out of the 124 PriRef pts, 78 (62.9%) went on to receive subsequent lines of therapy. 39.7% of these PriRef pts received a mAb, such as daratumumab, elotuzumab or isatuximab, in the 2 nd or 3 rd line setting. When stratified by whether they received mAb therapy, median OS of PriRef pts who did not receive a mAb was 86.6 months, whereas median OS was not reached for the PriRef pts who received a mAb (p=0.98) (Figure 2). Conclusions: Our cohort analysis showed that early primary refractoriness to induction in newly diagnosed MM patients was not associated with a lower overall survival, despite lower utilization of triplet regimens and ASCT. Treatment with monoclonal antibodies in the 2 nd and 3 rd line setting may explain this finding. Since the approval of mAbs for myeloma, OS appears to be beneficially impacted. However, the effect of this class of therapy is yet to be fully appreciated in the real-world setting. Therefore, longer follow up data is needed to better assess the true impact of monoclonal antibodies on primary refractory multiple myeloma patients. Figure 1 Figure 1. Disclosures Neparidze: GlaxoSmithKline: Research Funding; Janssen: Research Funding; Eidos Therapeutics: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3598-3598 ◽  
Author(s):  
Jean-Luc Harousseau ◽  
Donna Weber ◽  
Meletios Dimopoulos ◽  
Marta Olesnyckyj ◽  
Zhinuan Yu ◽  
...  

Abstract Background: There is ongoing debate about what would be considered an acceptable response to therapy in patients with relapsed/refractory multiple myeloma (MM). Lenalidomide (Len) is an immunomodulatory compound. Two recent phase III randomised trials (MM-009 and MM-010) showed Len in combination with Dexamethasone (Dex) provided significantly better overall response (OR), complete response (CR), overall survival (OS) and time to progression (TTP) compared with Dex alone in patients with relapsed/refractory MM. Approximately 60% of patients achieved an OR (61% in MM-009 and 60% in MM-010), including a CR in 15% (14% and 16%, respectively), Median OS was ≥29.5 months (29.5 and not yet reached) and median TTP was ≥11.1 months (11.1 months and 11.3 months, respectively). Here, we compare outcomes in patients with a CR or near CR (nCR) vs those with a partial response (PR). Methods: Data for patients who received Len/Dex in the MM-009 or MM-010 studies were pooled for this analysis. Patients received Len (25 mg/day on days 1–21 of every 28-day cycle) and Dex 40mg PO q.d. on days 1–4, 9–12, and 17–20 (for the first four cycles). After 4 cycles, Dex 40 mg/day was administered only on days 1–4. Response to therapy, TTP, OS, duration of response, and time to response, as well as adverse events were assessed. Response rate and TTP are based on data obtained before unblinding (June 2005 [MM-009] and August 2005 [MM-010]). Results: Of 353 patients treated with Len/Dex, 214 (61%) had PR or better (86 with CR/nCR; 128 with PR). Baseline characteristics were comparable between patient groups; most patients had stage III disease (63% vs 66%) and an Eastern Cooperative Oncology Group (ECOG) score of 0–1 (83% vs 90%). More patients with CR/nCR had received ≥2 prior antimyeloma regimens/stem cell transplant than patients with PR (88% vs 70%). Patients with CR/nCR had significantly longer median TTP and OS compared with those with PR (Table). The median duration of response was significantly higher among patients with CR/nCR (not yet reached vs 38 weeks; p&lt;0.001). Fewer patients with CR/nCR relapsed compared with those with PR (38% vs 43%). The times to CR/nCR and PR were comparable. The most common grade 3–4 adverse events were neutropenia (43% vs 41%), thrombocytopenia (13% vs 16%), anaemia (12% vs 9%) and pneumonia (12% vs 6%). Rates of febrile neutropenia were low (1% vs 3%). Conclusion: Len/Dex treatment is associated with longer OS and TTP in patients with relapsed/refractory MM who achieve a CR than in patients who achieve a PR. Len/Dex CR/nCR (n=86) PR (n=128) P value Median OS (weeks) &gt;132.7 &gt;118.1 &lt;0.01 Median TTP (weeks) &gt;65.1 46.0 &lt;0.001 (n=84) (n=126) Duration of response (weeks) NE 38 &lt;0.001 Time to response (weeks) 10.2 10.9 NE = not yet evaluable


2012 ◽  
Vol 30 (24) ◽  
pp. 2946-2955 ◽  
Author(s):  
Pieter Sonneveld ◽  
Ingo G.H. Schmidt-Wolf ◽  
Bronno van der Holt ◽  
Laila el Jarari ◽  
Uta Bertsch ◽  
...  

Purpose We investigated whether bortezomib during induction and maintenance improves survival in newly diagnosed multiple myeloma (MM). Patients and Methods In all, 827 eligible patients with newly diagnosed symptomatic MM were randomly assigned to receive induction therapy with vincristine, doxorubicin, and dexamethasone (VAD) or bortezomib, doxorubicin, and dexamethasone (PAD) followed by high-dose melphalan and autologous stem-cell transplantation. Maintenance consisted of thalidomide 50 mg (VAD) once per day or bortezomib 1.3 mg/m2 (PAD) once every 2 weeks for 2 years. The primary analysis was progression-free survival (PFS) adjusted for International Staging System (ISS) stage. Results Complete response (CR), including near CR, was superior after PAD induction (15% v 31%; P < .001) and bortezomib maintenance (34% v 49%; P < .001). After a median follow-up of 41 months, PFS was superior in the PAD arm (median of 28 months v 35 months; hazard ratio [HR], 0.75; 95% CI, 0.62 to 0.90; P = .002). In multivariate analysis, overall survival (OS) was better in the PAD arm (HR, 0.77; 95% CI, 0.60 to 1.00; P = .049). In high-risk patients presenting with increased creatinine more than 2 mg/dL, bortezomib significantly improved PFS from a median of 13 months to 30 months (HR, 0.45; 95% CI, 0.26 to 0.78; P = .004) and OS from a median of 21 months to 54 months (HR, 0.33; 95% CI, 0.16 to 0.65; P < .001). A benefit was also observed in patients with deletion 17p13 (median PFS, 12 v 22 months; HR, 0.47; 95% CI, 0.26 to 0.86; P = .01; median OS, 24 months v not reached at 54 months; HR, 0.36; 95% CI, 0.18 to 0.74; P = .003). Conclusion Bortezomib during induction and maintenance improves CR and achieves superior PFS and OS.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1869-1869 ◽  
Author(s):  
Paul Richardson ◽  
Jeffrey Lee Wolf ◽  
Andrzej Jakubowiak ◽  
Jeffrey A. Zonder ◽  
Sagar Lonial ◽  
...  

Abstract Abstract 1869 Poster Board I-894 INTRODUCTION: Perifosine is an orally -bioavailable, novel signal transduction modulator with multiple pathway effects including inhibition of Akt and activation of JNK. We conducted a phase I/II study of perifosine + bortezomib (Vel) +/− dexamethasone (Dex) which demonstrated encouraging safety and clinical activity in 73 evaluable Vel relapsed and Vel refractory patients (pts) (ASH 2008 # 870). Herein, we report on the updated safety, overall response rate (ORR), time to progression (TTP) and overall survival (OS) results of perifosine + Vel (+/− Dex), in multiple myeloma (MM) pts with advanced disease and who were previously relapsed from and/or refractory to Vel. METHODS: The phase I stage of the study enrolled a total of 18 pts with a selected phase II dose of perifosine 50 mg qd + Vel 1.3 mg/m2 (d 1, 4, 8, 11) in 21-d cycles. The phase II enrolled 66 pts. Dex 20mg (on day of and after each Vel dose) could be added in pts with progressive disease (PD). NCI CTCAE v3.0 was used for toxicity assessment. Response was assessed by modified EBMT and Uniform criteria. RESULTS: A total of 84 pts have been enrolled comprised of 51 men and 33 women, median age 63 y (range 35 – 89): 83% of pts had relapsed/refractory MM, with a median of 5 lines of prior treatment (range 1–13). Prior therapy included Vel (100%), dex (98%), lenalidomide (75%), thalidomide (74%) and SCT (57%), with pts receiving a median of 2 prior Vel-based treatments. Seventy three pts were evaluable for response (having completed at least two or more cycles of therapy), and all 84 were assessed for safety. Most common (≥ 10%) grade 1 / 2 events included nausea, diarrhea, vomiting, fatigue and anorexia, which were manageable with supportive care and dose reductions. Grade 3 / 4 adverse events ≥ 5% included thrombocytopenia, neutropenia, anemia, hyponatremia and diarrhea. No DVT has been seen, and only 1 worsening peripheral neuropathy from grade 1 to 3 was reported, which proved reversible after treatment discontinuation. Two pts had perifosine reduced to 50 mg (GI related) in the phase 1 and 5 pts reduced to qod dosing in the phase II: 15 pts had Vel dose reductions primarily due to hematologic toxicity. No treatment related mortality was seen. Of the 11 pts who were inevaluable for efficacy, 6 were due to toxicity resulting in failure to complete two cycles or more of therapy; 3 for withdrawal of consent/ non-compliance, and 2 due to rapid disease progression within 1 cycle. ORR, TTP, and OS are as follows: As of August 2009, the median OS for all evaluable study pts and Vel relapsed pts has not been reached with 40/73 (55%) pts and 14/20 (70%) pts alive, respectively. 26/53 (49%) Vel refractory pts remain alive (range 9 mos to 3 years). The median OS for the 60/73 pts (82%) with stable disease or better is currently 33.4 mos. CONCLUSIONS: Perifosine in combination with Vel (+/− dex) has been generally well tolerated and continues to demonstrate impressive activity in both heavily pre-treated, Vel-refractory pts, with an ORR of 38% and OS of 16 mos, and in Vel-relapsed pts with an ORR of 55%, a median TTP of 8.8 months and a median OS of 25 mos, to date. Moreover, 80% of pts achieved stable disease or better, with a median OS of 33.4 mos. A phase III randomized, trial evaluating perifosine vs. placebo when combined with Vel and Dex in Vel-exposed pts with relapsed and refractory MM has been granted special protocol assessment (SPA) and is expected to commence by the end of the year. Disclosures: Richardson: Keryx Biopharmaceuticals: Honoraria. Gardner:Keryx Biopharmaceuticals: Employment, Equity Ownership. Sportelli:Keryx Biopharmaceuticals: Employment, Equity Ownership. Anderson:Keryx Biopharmaceuticals: Consultancy.


2021 ◽  
Vol 34 (Supplement_1) ◽  
Author(s):  
Feng Wang ◽  
Qingxia Fan ◽  
Junsheng Wang ◽  
Tao Wu ◽  
Yonggui Hong ◽  
...  

Abstract   Esophageal squamous cell carcinoma (ESCC) as a common malignancy is prevalent in East Asia and in eastern and southern Africa. Although pembrolizumab, nivolumab and camrelizumab are respectively recommended as second-line treatment for advanced ESCC due to improved overall survival (OS), objective response rate (ORR) was modest. New effective treatments are needed. Hence, the study of camrelizumab plus apatinib (VEGFR2 inhibitor) as second-line treatment for advanced ESCC was performed. Methods This ongoing phase II trial (NCT03736863) in six sites in China enrolled pts aged 18-75 with unresectable locally advanced, locally recurrent, or metastatic ESCC that progressed or were intolerant after first-line chemotherapy, and an ECOG performance status of 0-1. Pts received 200 mg camrelizumab intravenously every 2 weeks and apatinib 250 mg orally once per day in 4-week cycles until disease progression, unacceptable adverse events (AEs) or withdrawal of consent. The primary endpoint was investigator-assessed ORR. Secondary endpoints included disease control rate (DCR), progression-free survival (PFS) and OS. Results At data cutoff (Feb 28, 2021), 52 pts were enrolled, including 42 males and 50 with distant metastases, with the median age of 62 years. In the evaluable population of 39 pts, ORR without confirmation was 43.59% and DCR was 94.87%. The median duration of response was 6.9 months (95% CI 4.57–9.23). The median PFS was 6.8 month (95% CI 2.66–10.94). The 12-month overall survival was 52.2%. A total of 80.8% of pts had treatment-related AEs (TRAEs) with 46.2% of grade ≥ 3 TRAEs. The safety profile of camrelizumab and apatinib was consistent with other anti–PD-1 antibodies and angiogenesis inhibitors. Conclusion This is the first study that evaluates the combination anti–PD-1 antibody and anti-angiogenesis inhibitor as a second-line therapy for advanced ESCC. Camrelizumab plus apatinib showed encouraging clinical efficacy and acceptable safety. Further phase III randomized trials are warranted.


2021 ◽  
Vol 34 (Supplement_1) ◽  
Author(s):  
Yan Zheng ◽  
Jiangong Zhang ◽  
Wenqun Xing

Abstract   In recent years, immune checkpoint inhibitors (ICIs) have shown promising results in the treatment of ESCC. More than 20 phase II clinical trials have been launched to explore combinations of ICIs in the neoadjuvant setting for ESCC. Based on our phase II clinical trial, a two-arm phase III trial was launched in our Hospital. Methods A two-arm phase III trial was launched in April 2020 in our Hospital. Patient recruitment will be completed within 18 months. The primary endpoint is event-free survival (EFS). The secondary endpoints include pathologic complete response (pCR), disease-free survival (DFS) rate, overall response rate (ORR), R0 resection rate, major pathologic response (MPR), adverse events (AEs), complication rate and quality of life (QOL). A biobank of pretreatment, resected tumor tissue and paired blood samples will be built for translational research in the future. Results Until Dec. 2021, one hundred and twenty ESCC patients recruited in the trial. The trial is ongoing. Conclusion This RCT directly compares NAC with neoadjuvant toripalimab plus chemotherapy in terms of EFS for locally advanced ESCC. The results may usher in a new era of resectable ESCC treatment.


Sign in / Sign up

Export Citation Format

Share Document