Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Clin Cancer Res ; 27(5): 1247-1255, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33115815

ABSTRACT

PURPOSE: Doublets of everolimus with letrozole or trastuzumab have demonstrated activity against HER2-positive breast cancer, suggesting that the triple combination can have synergistic anticancer activity. PATIENTS AND METHODS: This first-in-human dose-escalation study (NCT02152943) enrolled patients with hormone receptor- positive, HER2-positive (defined by amplification, overexpression, or mutation) treatment-refractory advanced cancers to receive escalating doses (3+3 design) of daily oral letrozole (days 1-21), daily oral everolimus (days 1-21), and intravenous trastuzumab (day 1) every 21 days to determine dose-limiting toxicities (DLT) and MTD or recommended phase II dose (RP2D). RESULTS: A total of 32 patients with hormone receptor-positive, HER2-positive (amplification, n = 27; overexpression, n = 1; and mutation, n = 4) advanced breast cancer (n = 26) or other cancers (n = 6) were enrolled. The most frequent grade ≥3 adverse events included hyperglycemia (n = 4), anemia (n = 3), thrombocytopenia (n = 2), and mucositis (n = 2). DLTs included grade 3 mucositis and grade 4 neutropenia, and trastuzumab given as an 8 mg/kg loading dose on day 1 of cycle 1 followed by a 6 mg/kg maintenance dose on day 1 of subsequent cycles plus 10 mg everolimus daily and 2.5 mg letrozole daily every 21 days was declared as RP2D. Five patients with breast cancer (four with HER2 amplification and one with HER2 mutation) had partial responses. HER2 amplification in circulating cell-free DNA at baseline was associated with shorter progression-free and overall survival durations (P < 0.05). CONCLUSIONS: Everolimus, letrozole, and trastuzumab have a favorable safety profile and elicit encouraging signals of anticancer activity in patients with heavily pretreated hormone receptor- and HER2-positive advanced cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Neoplasms/drug therapy , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Adult , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Everolimus/administration & dosage , Female , Follow-Up Studies , Humans , Letrozole/administration & dosage , Male , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology , Non-Randomized Controlled Trials as Topic , Prognosis , Receptor, ErbB-2 , Retrospective Studies , Survival Rate , Trastuzumab/administration & dosage
2.
Clin Cancer Res ; 26(21): 5579-5587, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33055173

ABSTRACT

PURPOSE: Preclinical and early clinical data suggested that combining histone deacetylase (HDAC) and mTOR inhibitors can synergistically inhibit Hodgkin lymphoma. PATIENTS AND METHODS: During the dose-escalation study (ClinicalTrials.gov number: NCT01087554) with the HDAC inhibitor vorinostat and the mTOR inhibitor sirolimus (V+S), a patient with Hodgkin lymphoma refractory to nine prior therapies demonstrated a partial response (PR) lasting for 18.5 months, which promoted additional enrollment of patients with Hodgkin lymphoma as well as exploration of an alternative combination of vorinostat and mTOR inhibitor everolimus (V+E). RESULTS: A total of 40 patients with refractory Hodgkin lymphoma received V+S (n = 22) or V+E (n = 18). Patients received a median of five prior therapies, including brentuximab (n = 39), autologous stem cell transplantation (n = 26), and allogeneic stem cell transplantation (n = 12). The most frequent grade ≥3 treatment-related adverse event was thrombocytopenia in 55% and 67% of patients treated with V+S and V+E, respectively. Complete response was reported in 6 (27%) patients treated with V+S and 2 (11%) patients treated with V+E, and PR was reported in 6 patients (27%) treated with V+S and 4 (22%) patients treated with V+E (objective response rate of 55% and 33%, respectively). In summary, combined HDAC and mTOR inhibition had encouraging activity in heavily pretreated patients with relapsed/refractory Hodgkin lymphoma and warrants further investigation. CONCLUSIONS: Combined HDAC and mTOR inhibition has salutary activity in patients with relapsed refractory Hodgkin lymphoma and warrants further investigation.


Subject(s)
Everolimus/administration & dosage , Histone Deacetylases/genetics , Hodgkin Disease/drug therapy , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/genetics , Vorinostat/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols , Brentuximab Vedotin/administration & dosage , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Everolimus/adverse effects , Female , Hematopoietic Stem Cell Transplantation , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/adverse effects , Humans , Male , Middle Aged , Recurrence , Sirolimus/adverse effects , Stem Cell Transplantation , TOR Serine-Threonine Kinases/antagonists & inhibitors , Vorinostat/adverse effects , Young Adult
3.
Invest New Drugs ; 37(4): 738-747, 2019 08.
Article in English | MEDLINE | ID: mdl-30993587

ABSTRACT

Purpose Brentuximab vedotin (BV) is an anti-CD30 antibody-drug conjugate used in the treatment of several types of lymphomas. Expression of the target antigen has also been reported on a variety of malignant tumors of nonlymphoid origin. This phase 2, open-label study evaluated the safety and antitumor activity of BV in patients with CD30-expressing nonlymphomatous malignancies. Methods Patients were dosed with 1.8 or 2.4 mg/kg BV once every three weeks. Antitumor activity was assessed at Cycles 2, 4, and every 4 cycles thereafter. Patients with stable disease or better were eligible to continue treatment until disease progression, unacceptable toxicity, or study closure. Results Of the 2693 patients screened, 3.8% had solid tumors with CD30 expression and 63 eligible patients with solid tumors enrolled in this study. The most common CD30 positive solid tumors were testicular cancer and mesothelioma. Both subtypes had more than one patient with an objective response. The median duration of BV exposure was 6.1 weeks. The disease control rate, defined as achieving stable disease or better at any point during the study, was 55%. The objective response rate was 11%, with a median duration of response of 2.92 months. The most common adverse events reported were fatigue (57%), nausea (33%), and decreased appetite (32%). Conclusion The safety profile of BV in patients with solid tumors was similar to the known safety profile of BV. In solid tumors, BV had modest activity as a single agent, which was similar to other second-line treatments already available to patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Brentuximab Vedotin/therapeutic use , Ki-1 Antigen/metabolism , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/adverse effects , Brentuximab Vedotin/adverse effects , Female , Humans , Male , Middle Aged , Neoplasms/metabolism , Treatment Outcome , Young Adult
4.
J Hematol Oncol ; 12(1): 145, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31888672

ABSTRACT

BACKGROUND: In 2007, we initiated IMPACT, a precision medicine program for patients referred for participation in early-phase clinical trials. We assessed the correlation of factors, including genomically matched therapy, with overall survival (OS). PATIENTS AND METHODS: We performed molecular profiling (Clinical Laboratory Improvement Amendments) (genes ≤ 182) for patients with lethal/refractory advanced cancers referred to the Phase 1 Clinical Trials Program. Matched therapy, if available, was selected on the basis of genomics. Clinical trials varied over time and included investigational drugs against various targets (single agents or combinations). Patients were followed up for up to 10 years. RESULTS: Of 3487 patients who underwent tumor molecular profiling, 1307 (37.5%) had ≥ 1 alteration and received therapy (matched, 711; unmatched, 596; median age, 57 years; 39% men). Most common tumors were gastrointestinal, gynecologic, breast, melanoma, and lung. Objective response rates were: matched 16.4%, unmatched 5.4% (p < .0001); objective response plus stable disease ≥ 6 months rates were: matched 35.3% and unmatched 20.3%, (p < .001). Respective median progression-free survival: 4.0 and 2.8 months (p < .0001); OS, 9.3 and 7.3 months; 3-year, 15% versus 7%; 10-year, 6% vs. 1% (p < .0001). Independent factors associated with shorter OS (multivariate analysis) were performance status > 1 (p < .001), liver metastases (p < .001), lactate dehydrogenase levels > upper limit of normal (p < .001), PI3K/AKT/mTOR pathway alterations (p < .001), and non-matched therapy (p < .001). The five independent factors predicting shorter OS were used to design a prognostic score. CONCLUSIONS: Matched targeted therapy was an independent factor predicting longer OS. A score to predict an individual patient's risk of death is proposed. TRIAL REGISTRATION: ClinicalTrials.gov, NCT00851032, date of registration February 25, 2009.


Subject(s)
Biomarkers, Tumor/genetics , Liver Neoplasms/mortality , Molecular Targeted Therapy , Neoplasms/mortality , Precision Medicine/methods , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/antagonists & inhibitors , Case-Control Studies , Clinical Trials, Phase I as Topic , Female , Follow-Up Studies , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/pathology , Prognosis , Protein Kinases/chemistry , Response Evaluation Criteria in Solid Tumors , Survival Rate , Young Adult
5.
Invest New Drugs ; 36(6): 1016-1025, 2018 12.
Article in English | MEDLINE | ID: mdl-29611022

ABSTRACT

Background We sought to determine the recommended phase II dose (RP2D) and schedule of GSK2141795, an oral pan-AKT kinase inhibitor. Patients and Methods Patients with solid tumors were enrolled in the dose-escalation phase. Pharmacokinetic (PK) analysis after a single dose (Cycle 0) informed dose escalation using accelerated dose titration. Once one grade 2 toxicity or dose-limiting toxicity was observed in Cycle 1, the accelerated dose titration was terminated and a 3 + 3 dose escalation was started. Continuous daily dosing was evaluated along with two intermittent regimens (7 days on/7 days off and 3 times per week). In the expansion phase at RP2D, patients with endometrial or prostate cancer, as well as those with select tumor types with a PIK3CA mutation, AKT mutation or PTEN loss, were enrolled. Patients were evaluated for adverse events (AEs), PK parameters, blood glucose and insulin levels, and tumor response. Results The RP2D of GSK2141795 for once-daily dosing is 75 mg. The most common (>10%) treatment-related AEs included diarrhea, fatigue, vomiting, and decreased appetite. Most AEs were low grade. The frequency of hyperglycemia increased with dose; however, at the RP2D, grade 3 hyperglycemia was only reported in 4% of patients and no grade 4 events were observed. PK characteristics were favorable, with a prolonged half-life and low peak-to-trough ratio. There were two partial responses at the RP2D in patients with either a PIK3CA mutation or PTEN loss. Conclusion GSK2141795 was safe and well-tolerated, with clinical activity seen as monotherapy at the RP2D of 75 mg daily. NCT00920257.


Subject(s)
Diamines/pharmacokinetics , Diamines/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Administration, Oral , Adult , Aged , Aged, 80 and over , Cohort Studies , Diamines/administration & dosage , Diamines/adverse effects , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Mutation/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/administration & dosage , Pyrazoles/adverse effects
6.
Article in English | MEDLINE | ID: mdl-32913996

ABSTRACT

PURPOSE: Comprehensive genomic profiling (CGP) detects several classes of genomic alterations across numerous genes simultaneously and can match more patients with genomically targeted therapies than conventional molecular profiling. The current study estimated the costs of anticancer drugs and overall survival (OS) for patients who were treated with matched and unmatched therapy. METHODS: Costs were estimated for patients with complete data (188 of 500 patients) from a prospective, nonrandomized study of patients with diverse refractory cancers who underwent CGP and were treated with matched or unmatched therapy. We assessed mean time to treatment failure (TTF) and mean observed OS. Patient-specific drug and administration costs were imputed for the first regimen after CGP on the basis of drug classes, unit costs, and time on treatment. RESULTS: Patients on matched (n = 122) versus unmatched (n = 66) therapy had longer mean TTF (+1.5 months) and observed OS (+2.4 months) and higher drug costs (+$38,065; all P < .01). Increased drug costs were largely attributable to the longer duration of therapy associated with extended TTF (66.3%) rather than higher monthly drug costs (33.7%). Incremental increases in TTF (+1.9 months v +1.2 months) and observed OS (+2.5 months v +2.1 months) between matched and unmatched therapies were larger for those who underwent CGP in earlier- versus later-line therapy. Incremental increases in drug costs between matched and unmatched therapies were lower for earlier- compared with later-line therapy (+$27,000 v +$43,000, respectively). CONCLUSION: Matched therapy was associated with longer TTF, increased OS, and manageable incremental cost increases compared with unmatched therapy. Most of these increased costs were a result of the longer duration of therapy rather than higher monthly drug costs. The benefits of matching were numerically greater in earlier versus later lines of therapy, which is consistent with the value of early use of CGP.

7.
Invest New Drugs ; 36(3): 416-423, 2018 06.
Article in English | MEDLINE | ID: mdl-29047029

ABSTRACT

Background Both MET and c-SRC are important mediators of cancer progression and there is cross talk between the two molecules. Preclinical studies have demonstrated combination of MET and c-SRC inhibitors is effective in multiple cancer types. Methods We analyzed the safety and efficacy of administering a c-SRC inhibitor (dasatinib) in combination with a MET inhibitor (crizotinib) in a two-arm concurrent phase I study. Arm A consisted of crizotinib fixed at 250 mg twice per day with escalation of dasatinib. Arm B consisted of dasatinib fixed at 140 mg daily with escalation of crizotinib. Endpoints included dose-limiting toxicities (DLTs), recommended phase II dose (RP2D), and response (RECIST 1.1). Results We enrolled 61 patients (arm A: 31, arm B: 30). The most common cancers were sarcoma (21%) and prostate cancer (16%). In Arm A, at dose level 2 (DL2), 40% (2/5) experienced DLTs. In the expanded DL1, 21% (4/19) experienced DLTs (all grade 3). In Arm B, at DL2, 50% (2/4) experienced DLTs. In the expanded DL1, 22% (4/18) experienced DLTs (all grade 3). RP2D was determined to be arm A, DL1 (250 mg crizotinib orally twice per day plus 50 mg dasatinib orally daily). Partial response (N = 1) and stable disease for ≥6 months (N = 3) were seen. Conclusions The combination of crizotinib and dasatinib is safe to administer but tolerability is limited given the high rate of adverse events. Responses and durable stable disease were limited. Further precision therapy approach using this specific combination may be difficult given the toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Crizotinib/therapeutic use , Dasatinib/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Crizotinib/adverse effects , Dasatinib/adverse effects , Female , Humans , Male , Middle Aged , Neoplasm Staging , Protein Kinase Inhibitors/adverse effects , Treatment Outcome
8.
Mol Cancer Ther ; 17(3): 671-676, 2018 03.
Article in English | MEDLINE | ID: mdl-29237802

ABSTRACT

Preclinical data suggest that combining a checkpoint inhibition with immunomodulatory derivative can increase anticancer response. We designed a dose-escalation study using a 3 + 3 design to determine the safety, maximum tolerated dose (MTD) or recommended phase II dose (R2PD) and dose-limiting toxicities (DLT) of the anti-CTLA-4 antibody ipilimumab (1.5-3 mg/kg intravenously every 28 days × 4) and lenalidomide (10-25 mg orally daily for 21 of 28 days until disease progression or unacceptable toxicity) in advanced cancers. Total of 36 patients (Hodgkin lymphoma, 7; melanoma, 5; leiomyosarcoma, 4; renal cancer, 3; thyroid cancer, 3; other cancers, 14; median of 3 prior therapies) were enrolled. The MTD has not been reached and ipilimumab 3 mg/kg and lenalidomide 25 mg have been declared as R2PD. DLT were grade (G) 3 rash (3 patients) and G3 pancreatitis (1 patient). G3/4 drug-related toxicities other than DLT were G3 anemia (5 patients), G3 thromboembolism (2 patients), G3 thrombocytopenia, G3 rash, G3 hypopituitarism, G3 pneumonitis, G3 transaminitis, and G4 hypopituitarism (all in 1 patient). Eight patients had tumor shrinkage per immune-related response criteria (-79% to -2%) including a PR (-79% for 7.2+ months) in a refractory Hodgkin lymphoma. Using comprehensive genomic profiling, a total mutation burden (mutations/Mb) was evaluated in 17 patients, with one of the patients achieving a PR demonstrated intermediate mutation burden. In conclusion, combination of ipilimumab and lenalidomide is well tolerated and demonstrated preliminary signals of activity in patients with refractory Hodgkin lymphoma and other advanced cancers. Mol Cancer Ther; 17(3); 671-6. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Adult , Aged , Anemia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , CTLA-4 Antigen/immunology , Dose-Response Relationship, Drug , Drug Administration Schedule , Exanthema/chemically induced , Female , Humans , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Ipilimumab/immunology , Lenalidomide/administration & dosage , Lenalidomide/adverse effects , Male , Middle Aged , Neoplasms/classification , Neoplasms/genetics , Young Adult
9.
Sci Rep ; 7(1): 15963, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29162825

ABSTRACT

Pazopanib is US FDA approved for the treatment of advanced soft tissue sarcomas. All patients with this disease ultimately develop resistance to therapy. Mechanisms of resistance include activation of the mTOR, histone deacetylase (HDAC), MAPK, and ERBB4 pathways. We hypothesized that combining pazopanib with other targeted agents inhibiting these pathways would increase response rates. We retrospectively evaluated the safety and efficacy of pazopanib plus vorinostat, everolimus, lapatinib or trastuzumab, and MEK inhibitor in patients with advanced sarcoma. The Cancer Geneome Atlas (TCGA) data was analyzed for HDAC, PI3K, HER2, and MAPK/RAS/RAF gene alterations from sarcoma TCGA. Of the 44 advanced sarcoma patients in these trials, 27 (61%) were male; 18 (41%) had bone sarcoma, and 26 (59%) had soft tissue sarcoma. Best response was partial response (PR) in four patients [(overall response rate (ORR) = 9%, 95% confidence interval [CI] 3% to 22%)]. The median progression-free survival (PFS) for all patients was 9.6 weeks (95% CI 8.0 to 15.7 weeks). Analysis of TCGA data revealed HDAC, PI3K, HER2, and MAPK/RAS/RAF gene alterations in 112/243 (46%) of patients predominantly HDAC1-11 (41%) alterations. Pazopanib combinations did demonstrate safety in combination with other agents. TCGA data suggests further evaluation of epigenetic pathway inhibitors in sarcoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Sarcoma/drug therapy , Sulfonamides/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adolescent , Adult , Aged , Clinical Trials as Topic , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Indazoles , Kaplan-Meier Estimate , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Prognosis , Receptor, ErbB-2/metabolism , Sarcoma/diagnostic imaging , Sarcoma/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
10.
Oncotarget ; 8(50): 87163-87173, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29152071

ABSTRACT

BACKGROUND: In 2011, we reported the outcomes of pancreatic cancer (PC) patients enrolled in phase I trials at our institution from 2004 through 2009. At the time, gemcitabine and erlotinib were the only Food and Drug Administration-approved drugs for PC and median overall survival (OS) from consultation in the phase I clinic was 5 months. We sought to determine the impact of novel therapeutics on PC patients in phase I trials. METHODS: We reviewed records of PC patients treated in phase I trials at our institution from January 2009 through December 2014. Survival was analyzed using the Kaplan-Meier method. RESULTS: Ninety-five patients were identified. The median age was 61 years (range, 40-84), 59% were men, and 41% had stage IV disease. The median OS from consultation in the phase I clinic was 5.8 months (95% confidence interval [CI], 4.5-6.8), and the 1-year OS rate was 9% (95% CI, 4%-17%). Three patients had partial responses and 18 had stable disease ≥ 4 months. CONCLUSION: We observed no improvement in OS between PC patients enrolled in phase I trials in 2004-2009 and 2009-2015. To substantially improve OS in this challenging disease, improved patient selection and science-driven, innovative trial designs will be key.

11.
JCO Precis Oncol ; 20172017.
Article in English | MEDLINE | ID: mdl-29082359

ABSTRACT

PURPOSE: Genomic profiling is increasingly used in the management of cancer. We have previously reported preliminary results of our precision medicine program. Here, we present response and survival outcomes for 637 additional patients who were referred for phase I trials and were treated with matched targeted therapy (MTT) when available. PATIENTS AND METHODS: Patients with advanced cancer who underwent tumor genomic analyses were treated with MTT when available. RESULTS: Overall, 1,179 (82.1%) of 1,436 patients had one or more alterations (median age, 59.7 years; men, 41.2%); 637 had one or more actionable aberrations and were treated with MTT (n = 390) or non-MTT (n = 247). Patients who were treated with MTT had higher rates of complete and partial response (11% v 5%; P = .0099), longer failure-free survival (FFS; 3.4 v 2.9 months; P = .0015), and longer overall survival (OS; 8.4 v 7.3 months; P = .041) than did unmatched patients. Two-month landmark analyses showed that, for MTT patients, FFS for responders versus nonresponders was 7.6 versus 4.3 months (P < .001) and OS was 23.4 versus 8.5 months (P < .001), whereas for non-MTT patients (responders v nonresponders), FFS was 6.6 versus 4.1 months (P = .001) and OS was 15.2 versus 7.5 months (P = .43). Patients with phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase pathway alterations matched to PI3K/Akt/mammalian target of rapamycin axis inhibitors alone demonstrated outcomes comparable to unmatched patients. CONCLUSION: Our results support the use of genomic matching. Subset analyses indicate that matching patients who harbor a PI3K and mitogen-activated protein kinase pathway alteration to only a PI3K pathway inhibitor does not improve outcome. We have initiated IMPACT2, a randomized trial to compare treatment with and without genomic selection.

12.
Eur J Cancer ; 83: 229-236, 2017 09.
Article in English | MEDLINE | ID: mdl-28755607

ABSTRACT

BACKGROUND: Cabozantinib (XL184), an orally bioavailable inhibitor of vascular endothelial growth factor receptor 2 and MET, was assessed in a cohort of ovarian carcinoma patients as part of a phase 2 randomised discontinuation trial (RDT) with cohorts from nine different tumour types. PATIENTS AND METHODS: Patients received 100-mg cabozantinib daily. Those with stable disease (SD) per Response Evaluation Criteria in Solid Tumors at week 12 were randomised to cabozantinib or placebo. Primary end-points were objective response rate (ORR) at week 12 and progression-free survival (PFS) after random assignment. RESULTS: Seventy patients with ovarian carcinoma, 50% of whom were platinum refractory/resistant, were enrolled in this RDT. Median PFS from day 1 was 5.5 months for all patients. The ORR at week 12 was 21%; one patient achieved a complete response (CR), and 14 patients (20%) achieved a confirmed partial response (PR). The overall disease control rate (CR + PR + SD) at week 12 was 50%. Throughout the study, 70% of the patients with ≥1 postbaseline scan had tumour regression, and randomisation was discontinued early. For patients with SD randomised to cabozantinib, PFS was 5.9 months after randomisation. The most common grade 3/4 adverse events were diarrhoea (14%), palmar-plantar erythrodysesthesia syndrome (6%), asthenia (6%), hypertension (6%) and neutropenia (6%). Dose reductions were required in 37% of the patients during the first 12 weeks. CONCLUSION: Cabozantinib demonstrates clinical activity, with acceptable toxicities, in patients with ovarian carcinoma based on ORR and regression of tumour target lesions. REGISTRATION: This trial is registered at ClinicalTrial.gov (NCT00940225).


Subject(s)
Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Ovarian Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Adult , Aged , Female , Humans , Middle Aged , Survival Analysis
13.
Oncologist ; 22(4): 377-e37, 2017 04.
Article in English | MEDLINE | ID: mdl-28275119

ABSTRACT

LESSONS LEARNED: Despite involvement of PI3K pathway activation in tumorigenesis of solid tumors, single-agent PI3K inhibitors have shown modest clinical activity.Preclinical evidence suggests that combining PI3K pathway inhibitors and chemotherapy can enhance antitumor effects.In patients with solid tumors, the PI3K inhibitor pilaralisib had a favorable safety profile but did not enhance the antitumor activity of paclitaxel plus carboplatin.Further clinical evaluation is warranted to identify effective combination strategies with PI3K pathway inhibitors. BACKGROUND: Pilaralisib (SAR245408) is an oral, pan-class I phosphoinositide 3-kinase (PI3K) inhibitor. This phase I dose-escalation study evaluated the maximum tolerated dose (MTD), safety, pharmacokinetics (PK), and pharmacodynamics of pilaralisib in capsule and tablet formulations, administered in combination with paclitaxel and carboplatin in patients with advanced solid tumors. METHODS: A 3 + 3 design was used. Pilaralisib was administered once daily (QD); paclitaxel (up to 175 mg/m2) and carboplatin (up to area under the curve [AUC] of 6) were administered on day 1 of 21-day cycles. An MTD expansion cohort of patients with endometrial carcinoma was included. RESULTS: Fifty-eight patients were enrolled. Six patients (10.3%) had dose-limiting toxicities, of which only rash (two patients, 3.4%) occurred in more than one patient. The MTD of pilaralisib tablets in combination with paclitaxel and carboplatin was determined to be 200 mg QD. The most frequently reported adverse events (AEs) of any grade were neutropenia (67.2%) and thrombocytopenia (67.2%). PK data showed no interaction between pilaralisib and paclitaxel/carboplatin. Tumor tissue showed moderate inhibition of PI3K and mitogen-activated protein kinase (MAPK) pathways. Seven of 52 evaluable patients had a partial response (PR; 13.5%). CONCLUSION: Pilaralisib had a favorable safety profile but did not enhance the antitumor activity of paclitaxel plus carboplatin in solid tumors. The Oncologist 2017;22:377-378.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/administration & dosage , Quinoxalines/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Aged, 80 and over , Carboplatin/administration & dosage , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Middle Aged , Neoplasms/pathology , Paclitaxel/administration & dosage , Protein Kinase Inhibitors/adverse effects , Quinoxalines/adverse effects , Sulfonamides/adverse effects
14.
Cancer Discov ; 7(4): 400-409, 2017 04.
Article in English | MEDLINE | ID: mdl-28183697

ABSTRACT

Entrectinib, a potent oral inhibitor of the tyrosine kinases TRKA/B/C, ROS1, and ALK, was evaluated in two phase I studies in patients with advanced or metastatic solid tumors, including patients with active central nervous system (CNS) disease. Here, we summarize the overall safety and report the antitumor activity of entrectinib in a cohort of patients with tumors harboring NTRK1/2/3, ROS1, or ALK gene fusions, naïve to prior TKI treatment targeting the specific gene, and who were treated at doses that achieved therapeutic exposures consistent with the recommended phase II dose. Entrectinib was well tolerated, with predominantly Grades 1/2 adverse events that were reversible with dose modification. Responses were observed in non-small cell lung cancer, colorectal cancer, mammary analogue secretory carcinoma, melanoma, and renal cell carcinoma, as early as 4 weeks after starting treatment and lasting as long as >2 years. Notably, a complete CNS response was achieved in a patient with SQSTM1-NTRK1-rearranged lung cancer.Significance: Gene fusions of NTRK1/2/3, ROS1, and ALK (encoding TRKA/B/C, ROS1, and ALK, respectively) lead to constitutive activation of oncogenic pathways. Entrectinib was shown to be well tolerated and active against those gene fusions in solid tumors, including in patients with primary or secondary CNS disease. Cancer Discov; 7(4); 400-9. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 339.


Subject(s)
Benzamides/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Colorectal Neoplasms/drug therapy , Indazoles/administration & dosage , Mammary Analogue Secretory Carcinoma/drug therapy , Melanoma/drug therapy , Oncogene Proteins, Fusion/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase , Benzamides/adverse effects , Benzamides/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Crizotinib , Dose-Response Relationship, Drug , Female , Humans , Indazoles/adverse effects , Indazoles/pharmacokinetics , Male , Mammary Analogue Secretory Carcinoma/genetics , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Middle Aged , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/genetics , Sequestosome-1 Protein/genetics
15.
Int J Cancer ; 140(1): 208-215, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27599876

ABSTRACT

Patients ≥65 years with cancer remain underrepresented in clinical trials, particularly in phase I clinical trials. We analyzed the clinical course of patients ≥65 years treated on phase I clinical trials with particular emphasis on toxicities. We identified 347 consecutive patients ≥65 years with advanced cancer in our phase I clinic from 01/2004-12/2009 and analyzed disease characteristics, toxicities, survival and response. Overall, 251 patients received a targeted agent, of whom 241 (96%) received an investigational, non-FDA-approved drug. Clinical benefit (complete response + partial response + stable disease ≥ 6 months) was noted in 61 patients (18%). Eighty-nine patients (26%) had grade 3/4 toxicity, commonly hematologic, including 6 dose-limiting toxicities and 1 treatment-related death (<0.01%). Median overall survival from first Phase I Clinic visit was 8.8 months (95% CI: 7.8-10.6); median time to treatment failure was 1.9 months (95% CI: 1.8-2.1). Multivariable analyses revealed 4 indicators of lack of clinical benefit (liver metastases, performance status [PS] >1, prior radiation, ≥5 prior treatments; p <0.0001). Patients age 70-79 years had a greater risk of grade 3/4 toxicities when treated with combinations (≥2 drugs) compared to monotherapy (p = 0.006). Predictors of shorter time to treatment failure and overall survival included PS >1, thrombocytosis, >2 metastatic sites, and elevated lactate dehydrogenase (p <0.05). Our results suggest that phase I clinical trials are well tolerated in patients ≥65 years. Additionally, we identified risk factors that may facilitate patient selection for clinical trial participation.


Subject(s)
Antineoplastic Agents/adverse effects , Drugs, Investigational/adverse effects , Neoplasms/drug therapy , Aged , Aged, 80 and over , Clinical Trials, Phase I as Topic , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/statistics & numerical data , Female , Humans , Male , Neoplasm Metastasis , Neoplasms/pathology , Patient Selection , Risk Factors , Survival Analysis , Treatment Failure , Treatment Outcome
16.
Oncotarget ; 7(41): 67521-67531, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27589687

ABSTRACT

Preclinical models suggest that histone deacetylase (HDAC) and mammalian target of rapamycin (mTOR) inhibitors have synergistic anticancer activity. We designed a phase I study to determine the safety, maximum tolerated dose (MTD), recommended phase II dose (RP2D), and dose-limiting toxicities (DLTs) of combined mTOR inhibitor sirolimus (1 mg-5 mg PO daily) and HDAC inhibitor vorinostat (100 mg-400 mg PO daily) in patients with advanced cancer. Seventy patients were enrolled and 46 (66%) were evaluable for DLT assessment since they completed cycle 1 without dose modification unless they had DLT. DLTs comprised grade 4 thrombocytopenia (n = 6) and grade 3 mucositis (n = 1). Sirolimus 4 mg and vorinostat 300 mg was declared RP2D because MTD with sirolimus 5 mg caused significant thrombocytopenia. The grade 3 and 4 drug-related toxic effects (including DLTs) were thrombocytopenia (31%), neutropenia (8%), anemia (7%), fatigue (3%), mucositis (1%), diarrhea (1%), and hyperglycemia (1%). Of the 70 patients, 35 (50%) required dose interruption or modification and 61 were evaluable for response. Partial responses were observed in refractory Hodgkin lymphoma (-78%) and perivascular epithelioid tumor (-54%), and stable disease in hepatocellular carcinoma and fibromyxoid sarcoma. In conclusion, the combination of sirolimus and vorinostat was feasible, with thrombocytopenia as the main DLT. Preliminary anticancer activity was observed in patients with refractory Hodgkin lymphoma, perivascular epithelioid tumor, and hepatocellular carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adolescent , Adult , Aged , Female , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Sirolimus/administration & dosage , Sirolimus/adverse effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Vorinostat , Young Adult
17.
Oncoscience ; 3(5-6): 164-72, 2016.
Article in English | MEDLINE | ID: mdl-27489863

ABSTRACT

There is limited data on co-expression of FGFR/FGR amplifications and PI3K/ AKT/mTOR alterations in breast cancer. Tumors from patients with metastatic breast cancer referred to our Phase I Program were analyzed by next generation sequencing (NGS). Genomic libraries were selected for all exons of 236 (or 182) cancer-related genes sequenced to average depth of >500× in a CLIA laboratory (Foundation Medicine, Cambridge, MA, USA) and analyzed for all classes of genomic alterations. We report genomic profiles of 112 patients with metastatic breast cancer, median age 55 years (range, 27-78). Twenty-four patients (21%) had at least one amplified FGFR or FGF. Fifteen of the 24 patients (63%) also had an alteration in the PI3K/ AKT/mTOR pathway. There was no association between alterations in FGFR/FGF and PI3K/AKT/mTOR (P=0.49). Patients with simultaneous amplification in FGFR/FGF signaling and the PI3K/AKT/mTOR pathway had a higher rate of SD≥6 months/PR/ CR when treated with therapies targeting the PI3K/AKT/mTOR pathway than patients with only alterations in the PI3K/AKT/mTOR pathway (73% vs. 34%; P=0.0376) and remained on treatment longer (6.8 vs. 3.7 months; P=0.053). Higher response rates were seen in patients with simultaneous amplification in FGFR/FGF signaling and alterations in the PI3K/AKT/mTOR pathway who were treated with inhibitors of that pathway.

18.
Mol Cancer Ther ; 15(10): 2475-2485, 2016 10.
Article in English | MEDLINE | ID: mdl-27466356

ABSTRACT

TP53 tumor-suppressor gene mutations are among the most frequent abnormalities in cancer, affecting approximately 40% of patients. Yet, there is no accepted way to target these alterations in the clinic. At the same time, antagonists of VEGFR or its ligand are best-selling oncology drugs, with multiple, expensive compounds approved. Although only a subset of patients benefit from these antiangiogenesis agents, no relevant biomarker has been identified. Interestingly, TP53 mutations upregulate VEGF-A and VEGFR2. We prospectively enrolled 500 patients, to be interrogated by comprehensive genomic profiling (CGP) (next-generation sequencing, 236 genes), and to be matched, whenever possible, with targeted agents. Herein, we analyze outcomes based on VEGF/VEGFR inhibitor treatment and presence of TP53 mutations. Of the 500 patients, 188 (37.6%; with ≥1 alteration) were treated; 106 (56% of 188) had tumors that harbored TP53 mutations. VEGF/VEGFR inhibitor therapy was independently associated with improvement in all outcome parameters [rate of stable disease (SD) ≥6 months/partial and complete remission (PR/CR); (31% versus 7%; TP53-mutant patients (who received no other molecular-matched agents) treated with versus without VEGF/VEGFR inhibitors), time-to-treatment failure, and overall survival (multivariate analysis: all P ≤ 0.01)] for the patients harboring TP53-mutant cancers, but improvement was not seen in any of these parameters for patients with TP53 wild-type neoplasms. We conclude that TP53 mutations predict sensitivity to VEGF/VEGFR inhibitors in the clinic. TP53 alterations may therefore be a ready biomarker for treatment with antiangiogenesis agents, a finding of seminal importance across the cancer field. Mol Cancer Ther; 15(10); 2475-85. ©2016 AACR.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adult , Aged , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm/genetics , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/mortality , Proportional Hazards Models , Protein Kinase Inhibitors/pharmacology , Receptors, Vascular Endothelial Growth Factor/genetics , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
19.
Cancer Chemother Pharmacol ; 78(1): 167-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27286994

ABSTRACT

PURPOSE: Tyrosine kinase inhibitor (TKI)-induced thyroid dysfunction has been identified as an important but manageable adverse effect of targeted therapy. Several studies have suggested that patients who develop hypothyroidism respond better to TKIs, but this relationship is not well elucidated. We evaluated the relationship between new-onset hypothyroidism and clinical response in patients with advanced cancers treated with TKIs at our institution. METHODS: We retrospectively reviewed records for patients from four clinical trials that included at least one TKI  therapy between January 2006 and December 2011. Patients with preexisting thyroid disease, including thyroid cancer, hypothyroidism, or hyperthyroidism, were excluded. Analysis of 197 patients was performed. Response was determined using RECIST 1.0. Clinical benefit was described as complete response, partial response, or stable disease greater than 4 months. Multivariable logistic regression analysis was performed to correlate patient characteristics with clinical response. RESULTS: The median age for the 197 patients was 58 years (range, 13-85 years), and 56 % were female. Of the 197 patients, 52 (26 %) developed hypothyroidism after therapy. Clinical benefit rates were 50 % in patients with new-onset hypothyroidism versus 34 % in patients without hypothyroidism. In the univariate model, the odds ratio (OR) for new-onset hypothyroidism was 1.9 [95 % confidence interval (CI) (1.0, 3.6) and p = 0.05]. We grouped tumor types into six categories (breast, colorectal carcinoma, melanoma, non-small cell lung cancer, pancreas, and other). When adjusted for tumor type, age (>50 years) and sex, the OR was 2.9 [95 % CI (1.3, 6.5) and p = 0.012] for new-onset hypothyroidism. CONCLUSION: New-onset hypothyroidism was associated with favorable clinical response in patients who received TKI treatment.


Subject(s)
Hypothyroidism/chemically induced , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Clinical Trials, Phase I as Topic , Female , Humans , Hypothyroidism/epidemiology , Logistic Models , Male , Middle Aged , Neoplasms/pathology , Protein Kinase Inhibitors/adverse effects , Retrospective Studies , Time Factors , Treatment Outcome , Young Adult
20.
Oncotarget ; 7(23): 35132-43, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27147567

ABSTRACT

High-grade epithelial ovarian cancer (HG-EOC) is the most lethal gynecologic malignancy worldwide Once patients develop chemoresistance, effective novel strategies are required to improve prognosis We analyzed characteristics and outcomes of 242 consecutive patients with HG-EOC participating in 94 phase I clinical trials at The University of Texas MD Anderson Cancer Center. Baseline lactate dehydrogenase levels, albumin levels, and number of metastatic sites were independent predictors of overall survival (OS). Receiving more than 1 phase I protocol was associated with improved OS (p < 0.001). Regimens including a chemotherapeutic agent plus bevacizumab or Aurora A kinase inhibitor led to a median progression-free survival (PFS) duration of more than 6 months. Although patients receiving bevacizumab-based regimens in the phase I clinical trials had significantly longer PFS than those receiving other anti-angiogenic therapies (p = 0.017), patients treated with vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) had significantly longer OS (12.2 months) than those not treated with VEGFR-TKIs (8.6 months, p = 0.015).In conclusion, anti-angiogenic therapy is one of the most important strategies for the treatment of HG-EOC, even in those who have already experienced tumor progression. Therefore, eligible patients with HG-EOC should be encouraged to participate in novel phase I studies of anti-angiogenic therapies, even after disease progression.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Carcinoma, Ovarian Epithelial , Clinical Trials, Phase I as Topic , Disease Progression , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasms, Glandular and Epithelial/mortality , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Proportional Hazards Models , Retrospective Studies , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...