scholarly journals Inhibition of the Ras/Raf/MEK/ERK and RET Kinase Pathways with the Combination of the Multikinase Inhibitor Sorafenib and the Farnesyltransferase Inhibitor Tipifarnib in Medullary and Differentiated Thyroid Malignancies

2011 ◽  
Vol 96 (4) ◽  
pp. 997-1005 ◽  
Author(s):  
David S. Hong ◽  
Maria E. Cabanillas ◽  
Jennifer Wheler ◽  
Aung Naing ◽  
Apostolia M. Tsimberidou ◽  
...  

Abstract Purpose: Ras/Raf/MAPK kinase/ERK and rearranged in transformation (RET) kinase pathways are important in thyroid cancer. We tested sorafenib, a B-Raf, RET, and vascular endothelial growth factor receptor kinase inhibitor, combined with tipifarnib, a farnesyltransferase inhibitor that inactivates Ras and other farnesylated proteins. Patients and Methods: We treated 35 patients with differentiated thyroid cancer (DTC) and medullary thyroid cancer (MTC) in a phase I trial. Sorafenib and tipifarnib were given for 21 d with 7 d rest in each 28-d cycle. Results: We enrolled 22 patients with metastatic DTC (16 papillary, five follicular, and one poorly differentiated) and 13 patients with MTC, of whom 15 with DTC and 10 with MTC reached first restaging. When tissue was available, eight of 15 DTC patients (53%) had B-Raf mutations; eight of 13 MTC (61.5%) patients had RET mutations. MTC partial response rate was 38% (five of 13) (duration = 9+, 12, 13, 16+, and 34+ months), stable disease of at least 6 months was 31% (four of 13). The DTC partial response rate was 4.5% (one of 22), and stable disease of at least 6 months was 36% (eight of 22). Median progression-free survival for all 35 patients was 18 months (95% confidence interval, 14.6 to not reached months). Median overall survival has not been reached, with a median follow-up of 24 months with 80% overall survival. Grade 1–2 toxicities were mainly rash, fatigue, and diarrhea. The most common grade 3–4 toxicities were rash, rise in amylase/lipase, and fatigue. Conclusions: Inhibiting the Ras/Raf/MAPK kinase/ERK and RET kinase pathways with sorafenib and tipifarnib is well tolerated and active against thyroid cancer.

2008 ◽  
Vol 26 (29) ◽  
pp. 4714-4719 ◽  
Author(s):  
Vandana Gupta-Abramson ◽  
Andrea B. Troxel ◽  
Anoma Nellore ◽  
Kanchan Puttaswamy ◽  
Maryann Redlinger ◽  
...  

PurposeGiven the molecular pathophysiology of thyroid cancer and the spectrum of kinases inhibited by sorafenib, including Raf kinase, vascular endothelial growth factor receptors, platelet-derived growth factor receptor, and RET tyrosine kinases, we conducted an open-label phase II trial to determine the efficacy of sorafenib in patients with advanced thyroid carcinoma.Patients and MethodsEligible patients with metastatic, iodine-refractory thyroid carcinoma received sorafenib 400 mg orally twice daily. Responses were measured radiographically every 2 to 3 months. The study end points included response rate, progression-free survival (PFS), and best response by Response Evaluation Criteria in Solid Tumors.ResultsThirty patients were entered onto the study and treated for a minimum of 16 weeks. Seven patients (23%; 95% CI, 0.10 to 0.42) had a partial response lasting 18+ to 84 weeks. Sixteen patients (53%; 95% CI, 0.34 to 0.72) had stable disease lasting 14 to 89+ weeks. Seventeen (95%) of 19 patients for whom serial thyroglobulin levels were available showed a marked and rapid response in thyroglobulin levels with a mean decrease of 70%. The median PFS was 79 weeks. Toxicity was consistent with other sorafenib trials, although a single patient died of liver failure that was likely treatment related.ConclusionSorafenib has clinically relevant antitumor activity in patients with metastatic, iodine-refractory thyroid carcinoma, with an overall clinical benefit rate (partial response + stable disease) of 77%, median PFS of 79 weeks, and an overall acceptable safety profile. These results represent a significant advance over chemotherapy in both response rate and PFS and support further investigation of this agent in these patients.


2021 ◽  
Vol 17 (1) ◽  
Author(s):  
Margaret L. Musser ◽  
Chad M. Johannes

Abstract Background Canine pancreatic carcinoma is a rare, aggressive tumour that is often diagnosed late in the course of disease. Effective treatment strategies have been elusive, and overall survival time is short. In humans, treatment with tyrosine kinase inhibitors alone, or in combination with IV gemcitabine, have been moderately effective. As canine and human pancreatic carcinomas share many clinical aspects, strategies that mimic human treatment regimens may confer a better outcome in canine patients. The aim of this study was to assess the role of the veterinary tyrosine kinase inhibitor, toceranib phosphate, in the treatment of cytologically or histologically confirmed canine pancreatic carcinomas. Results Retrospectively, medical records of dogs with confirmed pancreatic carcinoma treated with toceranib were reviewed. Eight dogs were identified that fit the inclusion criteria. Toceranib was well-tolerated by all patients. Six were treated in the gross disease setting. Four had image-based evaluation of clinical benefit (complete response, partial response, or stable disease of > 10 weeks). Of those patients, 1 achieved a partial response, 2 stable disease, and 1 had progressive disease, for an overall clinical benefit rate of 75 %. An additional dog had clinically stable disease that was not confirmed via imaging. The toceranib-specific median overall survival time was 89.5 days (range: 14–506 days). Conclusions Although limited in patient number, this small study suggests that toceranib may have biologic activity in dogs with pancreatic carcinoma. Larger, prospective studies are needed to confirm these preliminary results and define the use of toceranib in the microscopic disease setting.


2017 ◽  
Vol 35 (29) ◽  
pp. 3315-3321 ◽  
Author(s):  
Maria E. Cabanillas ◽  
Jonas A. de Souza ◽  
Susan Geyer ◽  
Lori J. Wirth ◽  
Michael E. Menefee ◽  
...  

Purpose Sorafenib and lenvatinib are oral multikinase inhibitors targeting vascular endothelial growth factor receptor (VEGFR) and approved for radioiodine (RAI)-refractory differentiated thyroid cancer (DTC). However, there are no approved second- or third-line therapies. MET is implicated in resistance to VEGFR inhibitors. Cabozantinib is an oral multikinase inhibitor targeting MET in addition to VEGFR and is approved for medullary thyroid cancer. In a phase I study of cabozantinib, five of eight patients with DTC previously treated with a VEGFR-targeted therapy had an objective response to cabozantinib. Patients and Methods Patients with RAI-refractory disease with Response Evaluation Criteria in Solid Tumor (RECIST) measurable disease and evidence of progression on prior VEGFR-targeted therapy were enrolled in this single-arm phase II study. The cabozantinib starting dose was 60 mg/day orally but could be escalated to 80 mg if the patient did not experience a response. Patients underwent tumor assessment according to RECIST v1.1 every 8 weeks. In this study, if at least five of 25 response-evaluable patients had an objective response, cabozantinib would be considered a promising agent in this patient population. Results Twenty-five patients were enrolled. The median age was 64 years, and 64% of patients were men. Twenty-one patients had received only one prior VEGFR-targeted therapy (sorafenib, pazopanib, or cediranib), and four patients had received two such therapies. The most common treatment-related adverse events were fatigue, weight loss, diarrhea, palmar-plantar erythrodysesthesia, and hypertension. One drug-related death was noted. Of the 25 patients, 10 (40%) had a partial response, 13 (52%) had stable disease, and two (8%) had nonevaluable disease. The median progression-free survival and overall survival were 12.7 months and 34.7 months, respectively. Conclusion Cabozantinib demonstrated clinically significant, durable objective response activity in patients with RAI-refractory DTC who experienced disease progression while taking prior VEGFR-targeted therapy.


2010 ◽  
Vol 20 (5) ◽  
pp. 787-793 ◽  
Author(s):  
Stephen A. Welch ◽  
Hal W. Hirte ◽  
Laurie Elit ◽  
Russel J. Schilder ◽  
Lisa Wang ◽  
...  

Objectives:Antiangiogenic strategies have demonstrated efficacy in epithelial ovarian cancer (EOC). Sorafenib is a novel multitargeted kinase inhibitor with antiangiogenic activity. Gemcitabine has known activity against EOC. A phase 1 clinical trial of this combination suggested activity in ovarian cancer with no dose-limiting toxicity. This phase 2 study was designed to examine the safety and efficacy of gemcitabine and sorafenib in patients with recurrent EOC.Methods:Patients with recurrent EOC after platinum-based chemotherapy and who had subsequently received up to 3 prior chemotherapy regimens were eligible. Gemcitabine (1000 mg/m2 intravenous [IV]) was administered weekly for 7 of 8 weeks in the first cycle, then weekly for 3 weeks of each subsequent 4-week cycle. Sorafenib (400 mg p.o. bid) was given continuously. The primary end point for this trial was objective response rate by the Response Evaluation Criteria in Solid Tumors. Secondary endpoints included Gynecologic Cancer Intergroup (GCIG) CA-125 response, time to progression, overall survival, and toxicity.Results:Forty-three patients were enrolled, and 33 completed at least 1 cycle. Two patients had a partial response (Response Evaluation Criteria in Solid Tumors objective response rate = 4.7%). Ten patients (23.3%) maintained response or stable disease for at least 6 months. GCIG CA-125 response was 27.9%. The median time to progression was 5.4 months, and the median overall survival was 13.0 months. Hematologic toxicity was common but manageable. The most common nonhematologic adverse events were hand-foot syndrome, fatigue, hypokalemia, and diarrhea.Conclusion:This trial of gemcitabine and sorafenib in recurrent EOC did not meet its primary efficacy end point, but the combination was associated with encouraging rates of prolonged stable disease and CA-125 response.


2004 ◽  
Vol 14 (5) ◽  
pp. 804-814 ◽  
Author(s):  
S. Tangjitgamol ◽  
H. T. See ◽  
S. Manusirivithaya ◽  
C. F. Levenback ◽  
D. M. Gershenson ◽  
...  

Ovarian carcinoma is a malignant disease with a high rate of recurrence, necessitating repeated chemotherapy treatments. We conducted a retrospective study in patients with platinum- and paclitaxel-resistant ovarian, fallopian tubes and primary peritoneal carcinoma patients treated at M.D. Anderson Cancer Center. We evaluated the responses, progression-free intervals, and overall survival duration of 51 patients after third-line chemotherapy treatment. The overall response rate was 16% (eight cases) with 2% complete response rate (one case) and 14% partial response rate (seven cases). Stable disease was achieved in 31% (16 cases). The progression-free intervals of 24 patients who had response and stable disease was 7.4 months (range, 1.4–18.4 months). The median overall survival of all patients was 15.8 months (95% CI, 8.1–23.4 months). The median survival duration of eight responders was not significantly different from that of 43 nonresponders, 18.9 months (95% CI, 2.4–35.4 months) versus 15.8 months (95% CI, 6.4–25.2 months), respectively (P = 0.73). In conclusion, third-line chemotherapy in our study results in a modest response and prolongation of progression-free interval without obvious impact on survival. The decision to utilize third-line chemotherapy will be a balance of the limited efficacy, toxicity of the agents, and the expertise of the clinician.


2019 ◽  
Vol 29 (5) ◽  
pp. 929-934 ◽  
Author(s):  
Meabh McNulty ◽  
Adarsh Das ◽  
Paul A Cohen ◽  
Andrew Dean

IntroductionResponse to neoadjuvant chemotherapy is measured by CT and the decision to proceed with interval surgery is made on the radiological response after two or three cycles of therapy. The Chemotherapy Response Score grades histological tumor regression in omental metastases resected at interval surgery and is associated with progression-free survival and overall survival. It is uncertain whether radiological response is associated with prognosis and whether radiological response predicts Chemotherapy Response Score.To assess if radiological response is associated with progression-free survival and overall survival. Additionally, to investigate whether radiological response predicts the Chemotherapy Response Score.MethodsRetrospective cohort study of patients with high-grade serous ovarian cancer treated with neoadjuvant chemotherapy. Radiological response was assessed by comparing CT imaging at baseline and after neoadjuvant chemotherapy using RECIST (Response Evaluation Criteria In Solid Tumors) and classified as stable disease, partial response, complete response, or progressive disease. Survival analysis was performed using Cox proportional-hazard models and the log-rank test.ResultsA total of 71 patients met the inclusion criteria. Of these, 51 had pre- and post-neoadjuvant chemotherapy CT scans available for analysis. Radiological response was not associated with progression-free survival or overall survival on univariate analysis (stable disease vs partial response; HR for progression-free survival 1.15; 95% CI 0.57 to 2.32; p = 0.690; HR for overall survival 1.19; 95% CI 0.57 to 2.46; p = 0.645). In a multivariate model, radiological response was not associated with either progression-free survival (stable disease vs partial response; HR=1.19; 95% CI 0.498 to 2.85; p = 0.694) or overall survival (stable disease vs partial response; HR=0.954; 95% CI 0.38 to 2.40; p = 0.920). There was a significant association between the Chemotherapy Response Score and radiological response (p = 0.005).DiscussionA partial response and stable disease on radiological assessment after neoadjuvant chemotherapy in women with advanced high-grade serous ovarian cancer were not associated with survival, despite having a correlation with the Chemotherapy Response Score.


2006 ◽  
Vol 24 (15) ◽  
pp. 2325-2331 ◽  
Author(s):  
Chandrajit P. Raut ◽  
Matthew Posner ◽  
Jayesh Desai ◽  
Jeffrey A. Morgan ◽  
Suzanne George ◽  
...  

PurposeWhile targeted inhibitors of tyrosine kinase activity demonstrate dramatic efficacy in the majority of patients with advanced gastrointestinal stromal tumors (GISTs), cure remains elusive and resistance to systemic therapy is a challenge. To assess the role of surgery in multimodality management of GISTs, we studied postoperative outcomes in patients treated with targeted kinase inhibitors for advanced GIST.MethodsWe evaluated outcomes in a single institution series of 69 consecutive patients who underwent surgery for advanced GISTs while receiving kinase inhibitors. Patients were categorized based on extent of disease before surgery (stable disease, limited disease progression, generalized disease progression) and surgical result (no evidence of disease, minimal residual disease, bulky residual disease).ResultsDisease status before surgery was associated with surgical result (P < .0001; median follow-up, 14.6 months). After surgery, there was no evidence of disease in 78%, 25%, and 7% of patients with stable disease, limited progression, and generalized progression, respectively. Bulky residual disease remained after surgery in 4%, 16%, and 43% of the patients with stable disease, limited progression, and generalized progression. Twelve-month progression-free survival was 80%, 33%, and 0% for patients with stable disease, limited progression, and generalized progression (P < .0001). Twelve-month overall survival was 95%, 86%, and 0% for patients with stable disease, limited progression, and generalized progression (P < .0001).ConclusionPatients with advanced GISTs exhibiting stable disease or limited progression on kinase inhibitor therapy have prolonged overall survival after debulking procedures. Surgery has little to offer in the setting of generalized progression.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1028-1028
Author(s):  
Stefania Paolini ◽  
Emanuela Ottaviani ◽  
Sarah Parisi ◽  
Federica Salmi ◽  
Barbara Lama ◽  
...  

Abstract Abstract 1028 Poster Board I-50 Background: Outcome of elderly acute myeloid leukemia (AML) patients is dismal. Targeted-therapies might improve current results by overcoming drug-resistance and reducing toxicity. In particular, the farnesyl-transferase inhibitor Tipifarnib (Zarnestra®), and the proteasome inhibitor Bortezomib (Velcade®), appeared synergistic in AML cells ex vivo, and their association was shown to be safe in vivo in a phase I trial by our group. Aim We conduced a phase II study aiming to assess efficacy and toxicity of Tipifarnib-Bortezomib association in AML patients >18 years, unfit for conventional therapy, or >60 years, in relapse. Furthermore, we aimed to identify biological features potentially predictive of clinical response. In particular, we focused on the RASGRP1/APTX ratio, which was previously found to be effective in predicting treatment response in patients treated with Tipifarnib alone. Methods: Bortezomib (1.0 mg/m2) was administered as weekly infusion for three consecutive weeks (days 1, 8, 15). Tipifarnib was administered at dose of 300-600 mg BID for 21 consecutive days. Response was assessed at the end of each cycle (28 days). Patients' withdrawn was planned in case of progression or stable disease after six cycles. Real-time quantitative-PCR (q-PCR) was used for RASGRP1/APTX quantification. Results: Eighty patients were enrolled (47 male). Median age was 71 years (43-89) and WBC at diagnosis was 4.2 × 109/L (0.5- 42.1). Thirty-two out of 80 patients had a secondary-AML, 14 had a high risk cytogenetic and 42 were previously untreated. Seventy-five patients actually initiated the treatment, 62 completed at least the first cycle while 13 early dropped out for non-leukemia related adverse event. Nine patients achieved complete remission (CR), 1 patients obtained a partial response (PR) and in 2 cases an hematological improvement (HI) was documented for an overall response rate (ORR) of 19%. Eighteen had progressive disease (PD) and the remaining showed stable disease (SD). Median time to response was 112 days, corresponding to 4 cycles (range 2-14). Marrow response (CR+PR) was significantly associated with overall survival (OS) (p<0.0001). RASGRP1/APTX was evaluated before treatment initiation on bone marrow (BM) and/or peripheral blood (PB). The median RASGRP/APTX value on BM was 15.3 (15-19.8) in responder patients and 2.2 (0.5-25.9) in non responders, respectively (p=0.00006). Its median value on PB was 31.6 (19.3-35.5) in responders and 6.4 (0.5-27.1) in non responders, respectively (p=0.00001). Interestingly, no marrow responses were recorded in patients with marrow RASGRP1/APTX ratio <8, while the response rate was 43% in patients with RASGRP1/APTX >8 (p<0.0001). Finally, RASGRP1/APTX levels significantly correlated with OS (p=0.001) with a median OS of 490 days and 162 days in patients with RASGRP1/APTX >8 and <8 respectively. Conversely, there was no correlation between cytogenetics, secondary AML, previous treatment and response or overall survival. Toxicity was overall mild, the most common adverse event being febrile neutropenia. Permanent treatment interruption due to Tipifarnib-Bortezomib related adverse events occurred in 13/75 (17%) of patients. With a median follow-up of 122 days (range 9-737), 57/75 (76%) patients are dead and 18/75 (24%) are alive, six of which in CR. Conclusion: We conclude that the clinical efficacy of the combination Tipifarnib-Bortezomib was similar to what reported for Tipifarnib alone. However, noteworthy, we could confirm that the RASGPR1/APTX BM or PB level is an effective predictor of response. Though higher RASGRP1/APTX is relatively rare (∼10% of cases), Tipifarnib (±Bortezomib) may represent an important option in a subset of high risk/frail AML patients. Acknowledgments: Supported by BolognAIL, AIRC, European LeukemiaNET, COFIN, FIRB 2006, Fondazione del Monte di Bologna e Ravenna. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 17133-17133 ◽  
Author(s):  
M. S. Boyar ◽  
M. Hesdorffer ◽  
R. N. Taub

17133 Background: A prior phase II trial of oxaliplatin and gemcitabine in malignant pleural mesothelioma reported activity with a 40% partial response rate and stable disease in 24% of 25 previously untreated patients (Schutte W. et al., Clin Lung Cancer 2003;4:294). This trial was designed to further evaluate the efficacy of oxaliplatin and gemcitabine in patients with malignant pleural or peritoneal mesothelioma. Methods: Patients with histologically-confirmed malignant pleural or peritoneal mesothelioma not amenable to curative surgical treatment are eligible for enrollment on this single-institution phase II trial. One prior chemotherapy regimen is allowed, including pemetrexed with a platinum compound or gemcitabine. Presence of at least one radiologically-measurable lesion that has not been previously irradiated is required. Gemcitabine 1000 mg/m2 IV over 90 minutes is administered followed by oxaliplatin 100 mg/m2 IV over 2 hours on day 1 of a 14-day cycle. Treatment is continued for at least 6 cycles unless unacceptable toxicity or disease progression occurs. The primary outcome is to evaluate efficacy as measured by tumor response rate. The sequential two-stage design allows for enrollment of a total of 29 patients if 3 of 18 patients enrolled in the first stage have at least a partial response. Results: Ten eligible patients have been enrolled thus far and 6 are evaluable for response. Sites of disease include pleural (3) and peritoneal (3), and histologic subtypes are epithelioid (5) and sarcomatoid (1). Four patients had received prior chemotherapy. A total of 42 cycles have been given with a median of 8 cycles per patient. There are no partial or complete responses. Four patients have had stable disease for at least 4 months. Two patients with stable disease had received prior pemetrexed/gemcitabine or pemetrexed/cisplatin. Frequently observed toxicities include grade II fatigue, neuropathy and upper gastrointestinal dysmotility. Conclusion: This regimen of gemcitabine and oxaliplatin may provide disease stabilization in a subset of patients with malignant pleural and peritoneal mesothelioma, including those who have received prior chemotherapy. The regimen is well-tolerated. This study remains open to accrual. No significant financial relationships to disclose.


2018 ◽  
Vol 36 (4_suppl) ◽  
pp. 450-450
Author(s):  
Nicha Wongjarupong ◽  
Mohamed Abdelrahim Muddathir Hassan ◽  
Cristobal T. Sanhueza ◽  
Mindy L. Hartgers ◽  
Fatima Hassan ◽  
...  

450 Background: The standard treatment for patients with gallbladder cancer is a combination of gemcitabine and cisplatin based on ABC-02 trial. However, there are no guidelines regarding treatment after first-line therapy. We retrospectively analyzed the efficacy and overall survival of different second-line regimens. Methods: We identified 203 patients with advanced gallbladder cancer who received palliative treatment between January 2000 and December 2015 at Mayo Clinic, Rochester. RECIST criteria was used to assess response. Results: 68 patients received second-line chemotherapy. Median age was 63 years (range: 32-86) and majority were males (60.6%). The median time from the diagnosis to the start of the second line chemotherapy was 8 (1-120) months. The most common used second-line chemotherapy were FOLFOX (14), gemcitabine alone (10), single agent fluoropyrimidine (11), gemcitabine with capecitabine (5), and capecitabine with oxaliplatin (4). There were 30 patients that received 5-fluorouracil based regimens, 20 patients received gemcitabine-based regimen, 3 patients received taxane-based regimen, and 15 patients received other types of chemotherapy. Median progression free survival and overall survival was 2.1 (1.8-2.7) and 16.7 (13.2-21.3) months respectively. There were 10 (52%), 11 (37%), 2 (67%), 5 (33%) with partial response and stable disease in 5-fluorouracil-based, gemcitabine-based, taxane-based, and others, respectively. There were no difference in PFS, with median PFS of 2.5, 2.0, 2.8 and 2.3 months, respectively (p=0.43). The overall survival were 15.7 (8.9-40.2), 15.0 (10.7-21.3), 40.3 (22.0-47.0), and 20.4 (9.2-30.7) months, respectively (p=0.83). There were 27 patients that received single agent chemotherapy and 41 patients that received combined regimen. There were 17 (42%) patients and 13 (48%) patients with partial response or stable disease in single and combined regimen. There were no differences in progression free survival and overall survival between single and multi agent chemotherapy. Conclusions: In this largest single institution study, second-line chemotherapy regimens for gallbladder cancer provided benefit in select patients and there is an urgent need to develop more active therapeutic regimens.


Sign in / Sign up

Export Citation Format

Share Document