A phase I pharmacokinetic study of a liposomal formulation of paclitaxel administered weekly to Asian patients with solid malignancies

2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 2581-2581 ◽  
Author(s):  
W. T. Lim ◽  
S. S. Leong ◽  
C. K. Toh ◽  
C. S. Ang ◽  
N. S. Wong ◽  
...  

2581 Background: Genexol-PM is a sterile, lyophilized polymeric micellar formulation of paclitaxel which is devoid of Cremophor EL and hence is more tolerable and less toxic. This phase 1 study sought to determine the maximum tolerated dose and the pharmacokinetic profile of Genexol-PM in Asian cancer patients with solid malignancies. Methods: Patients (N=35) refractory to previous chemotherapy were enrolled in a phase 1, open-label, dose-escalating study to assess safety, tolerability ad pharmacokinetics of Genexol-PM administered as a 1h infusion on a weekly basis for 3 weeks followed by a resting week. The starting dose was 80mg/m2. Cohorts of 1–6 patients were treated at 100, 120, 140, 160, 180 and the maximum administered dose was 200 mg/m2. Results: The median age was 56 years (range: 39 - 67 years) and two thirds of the enrolled patients were male (67%). Twenty-three patients (96%) had received prior chemotherapy, including eleven patients (46%) who had previously received taxane-based chemotherapy. The majority of patients had lung, nasopharyngeal and breast cancers. DLT was reached at 200 mg/m2. The MTD was 180 mg/m2. Grade 3 granulocytopenia was common in patients receiving Genexol-PM at doses of 120 mg/m2 or higher in the first cycle. The most common grade 3 non-haematologic adverse events in cycle 1 were fatigue, myalgia and neuropathy and occurred mainly at dose level 7 (200 mg/m2) in 4%, 4% and 8% of the patients. Five (21%) patients had partial response, 9 (38%) had stable disease and seven (29%) patients had disease progression. The pharmacokinetics of Genexol-PM displayed dose-proportionality, with both Cmax and AUC0-∞ values increasing by approximately 4- and 3-fold as the dose of Genexol-PM was increased from 80mg/m2 to 200mg/m2 with no significant change in clearance. The median total-body clearance of Genexol-PM for all patients was 43.9 L/hr. Conclusions: The weekly regimen of Genexol-PM was found to be well-tolerated and responses were observed in patients with refractory tumours, including patients who had failed taxane-based chemotherapy previously. No significant financial relationships to disclose.

2021 ◽  
Vol 11 (2) ◽  
Author(s):  
Jacob P. Laubach ◽  
Sascha A. Tuchman ◽  
Jacalyn M. Rosenblatt ◽  
Constantine S. Mitsiades ◽  
Kathleen Colson ◽  
...  

AbstractAdditional therapeutic options are needed for relapsed and refractory multiple myeloma (RRMM). We present data from a phase 1b, open-label, dose-escalation study (NCT01965353) of 20 patients with RRMM (median age: 63 years [range: 50–77]) and a median of four prior regimens (range: 2–14); 85% had refractory disease (lenalidomide [80%]; bortezomib [75%]; lenalidomide and bortezomib [50%]). Patients received a median of six cycles (range: 1–74) of panobinostat (10 or 15 mg), lenalidomide 15 mg, bortezomib 1 mg/m2, and dexamethasone 20 mg (pano-RVd). Median follow-up was ~14 months. Six dose-limiting toxicities were reported (mostly hematological); maximum tolerated dose of panobinostat (primary endpoint) was 10 mg. Most common adverse events (AEs) were diarrhea (60%) and peripheral neuropathy (60%); all grade 1/2. Grade 3/4 AEs occurred in 80% of patients and included decreased neutrophil (45%), platelet (25%) and white blood cell (25%) counts, anemia (25%) and hypophosphatemia (25%). No treatment-related discontinuations or mortality occurred. In evaluable patients (n = 18), overall response rate was 44%, and clinical benefit rate was 61%. Median duration of response was 9.2 months; progression-free survival was 7.4 months; overall survival was not reached. Pano-RVd proved generally well-tolerated and demonstrated potential to overcome lenalidomide and/or bortezomib resistance.


2020 ◽  
Vol 19 ◽  
pp. 153473542091144 ◽  
Author(s):  
Chunhoo Cheon ◽  
Seong-Gyu Ko

Background: Cancer is a major health problem worldwide and the leading cause of death in many countries. Preclinical studies have shown the therapeutic anticancer effects of SH003, a novel herbal medicine containing Astragalus membranaceus, Angelica gigas, and Trichosanthes kirilowii. The present study investigated the maximum tolerated dose of SH003 in patients with solid cancers. Methods: This open-label, dose-escalation trial used the traditional 3 + 3 dose-escalation design. Patients with solid cancers were recruited and administered 1 to 4 tablets of SH003 thrice daily for 3 weeks according to the dose level. Adverse events were evaluated according to the Common Terminology Criteria for Adverse Events (CTCAE). Dose-limiting toxicities (DLTs) were defined as Grade 3 or higher adverse events based on CTCAE. The maximum tolerated dose was defined as the highest dose at which no more than 1 of 6 patients experienced DLT. Results: The present study enrolled 11 patients. A total of 31 adverse events occurred. According to the CTCAE, all the observed adverse events were grade 2 or less and no adverse events of grade 3 or more corresponding to DLT occurred. Conclusion: The study results indicated that the maximum tolerated dose of SH003 was 4800 mg/day. A Phase 2 study is required to determine the efficacy of SH003 in patients with cancer at a dose of 4800 mg/day or less.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 14005-14005 ◽  
Author(s):  
T. J. Price ◽  
L. Lipton ◽  
J. Williams ◽  
J. McGreivy ◽  
S. McCoy ◽  
...  

14005 Background: AMG 706 is an oral, investigational multikinase inhibitor (MKI) with antiangiogenic and direct antitumor activity achieved by selectively targeting VEGF, PDGF and Kit receptors. Methods: This is a fully enrolled, phase 1b, open-label, dose- finding study. The objectives are to determine the maximum tolerated dose and to assess safety and PK of AMG 706 in pts with solid tumors receiving AMG 706 plus gemcitabine. Pts =18 years with ECOG 0–2 and no prior treatment with bevacizumab or VEGFr MKIs were assigned to cohorts receiving escalating doses of AMG 706 (50mg QD, 125mg QD or 75mg BID continuously from day 2 of cycle 1) plus gemcitabine (1000mg/m2 weekly for 7/8 wks, then 3/4 wks per cycle) for up to 11 cycles. Assessments include dose-limiting toxicities (DLT) (weeks 1–4) and tumor response (every 3 months). Results: 26 pts were enrolled and received at least 1 dose of AMG 706 (50mg QD n=11; 125mg QD n=6; 75mg BID n=9). All but 2 pts have completed the study. Median (range) age was 57 (25–77) yrs. 65% of pts received prior chemotherapy; 4 pts received prior gemcitabine (50mg QD n=2; 125mg QD n=1; 75mg BID n=1). There were 2 DLTs: grade 4 neutropenia (125mg QD), grade 3 deep vein thrombosis (75mg BID). Treatment-related adverse events (AE) occurring in = 10% of pts are shown in the table . The mean AMG 706 PK profiles were not markedly different when AMG 706 was dosed on the same day or 24 hours after gemcitabine administration. Objective tumor responses per RECIST for 26 evaluable pts were: 2 unconfirmed PR (50mg QD n=1; 125mg QD n=1), 7 SD (50mg QD n=3; 125mg QD n=1; 75mg BID n=3), 11 PD (50mg QD n=7; 125mg QD n=3; 75mg BID n=1), and 6 not available (125mg QD n=1; 75mg BID n=5). Conclusions: These preliminary data suggest that, in pts with solid tumors, AMG 706 combined with gemcitabine had an expected AE profile at the target once-daily dose of 125mg QD, with little effect on AMG 706 PK. The data provide a foundation for conducting further trials, potentially including biliary tumors. Final data will be presented. [Table: see text] No significant financial relationships to disclose.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e13513-e13513
Author(s):  
B. Besse ◽  
S. Almokadem ◽  
D. Planchard ◽  
I. Chico ◽  
C. L. Tsao ◽  
...  

e13513 Background: Volociximab is a chimeric monoclonal antibody that blocks fibronectin binding to α5β1 and induces apoptosis in proliferating endothelial cells. Its anti-angiogenic actions are independent of the VEGF pathway. This is the first clinical study of voloxicimab in patients with advanced NSCLC. Methods: This phase 1b multi-center open-label, dose-escalation study was designed to determine the maximum tolerated dose of V in combination with full doses of C (AUC=6mg/ml.min) P (200mg/m2) with cycles repeated every 3 wks for a maximum of 6 cycles followed by a maintenance treatment with V alone. Eligible pts had histologically confirmed untreated stage IIIb or IV NSCLC. In cohorts 1 and 2, pts received V at 10 mg/kg and 20mg/kg IV, respectively, on days 1 and 8, of the first 21 day cycle then every 21 days. In cohort 3, pts received V 30 mg/kg IV every 21 days from day 1. We present here the interim safety analysis and RECIST response data of the combination therapy. Results: A total of 22 patients (9, 6 and 7 in cohorts 1, 2 and 3 respectively) who received at least one dose of treatment constitute the safety population. Reported salient adverse events (any grade) were constipation (68%), asthenia (64%), nausea (59%), arthralgia (55%) and paresthesia (46%). Grade 3 bowel obstruction in one patient was considered a DLT in cohort 2. No DLT's were observed in cohort 3. Seven pts reported at least one serious adverse event (all Grade 3) including deep vein thrombosis (1), peripheral arterial occlusion (1), proteinuria (1), pneumonitis (1), small intestinal obstruction (1), pleural effusion (1), hypoxia (1), dehydration (1) and orthostatic hypotension (1). Partial response was seen in 6 pts and stable disease in 12 out of 18 pts who were evaluable for response by RECIST. Conclusions: The highest dose of V tested (30 mg/kg q3w) in combination with CP appears well tolerated and the regimen has promising clinical activity in advanced NSCLC. [Table: see text]


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 3562-3562
Author(s):  
S. Nallapareddy ◽  
D. Gustafson ◽  
S. Leong ◽  
W. Messersmith ◽  
J. Arnott ◽  
...  

3562 Background: ENMD-1198 (2-methoxyestra-1, 3, 5, (10) 16-tetraene-3-carboxamide), an analog of 2-methoxyestradiol (2ME2), has both antiangiogenic and antiproliferative effects in various tumor types. ENMD-1198 inhibits MT polymerization by binding to β-tubulin at the colchicine-binding site and inhibits HIF-1alpha. This Phase 1 study is evaluating the safety of ENMD-1198 to determine the maximum tolerated dose. Methods: Eligible pts had advanced cancer for which no effective therapy exists that is either evaluable by RECIST criteria or tumor markers that could be monitored for clinical benefit. Phase 1 dose escalation in 3+3 design for first 5 cohorts; modified to 1 pt cohorts for subsequent cohorts until Grade 2 treatment related toxicity, and then standard 3+3 design. All pts treated with once daily oral ENMD-1198 in 28-day cycles (with post-treatment drug-free observation period of 7–14 days in Cycle 1 only). Pts are treated until the appearance of significant treatment-emergent toxicities or disease progression (PD) occurs. Results: To date, 27 pts have been enrolled in 12 dose cohorts (range 5 to 550 mg/m2/d). Median age/performance status is 61/1. Total # of treatment cycles to date is 68, with a median of 2 cycles (range <1 to 15 cycles). Most frequent toxicities (all grades, n=22) were pain (77%), fatigue (55%), constipation (36%), neuropathy and nausea (both 32%), and anemia (27%). 4 pts have experienced stable disease (SD) for more than 2 cycles. There have been no objective responses to date. 2 pts experienced dose limiting toxicity with Grade 4 neutropenia in the 550 mg/m2/d cohort. Following drug holiday, pts restarted at 425 mg/m2/d and continued for at least 1–2 more cycles before being removed from study for PD. One pt (neuroendocrine ca pancreas) is experiencing prolonged SD at 60 mg/m2/d >14 cycles and a 2nd pt (prostate ca) experienced SD at 30mg/m2/d for 10 cycles. ENMD-1198 was absorbed rapidly after oral administration. There was a linear relationship between dose and drug exposure as measured by AUC across all doses (5 - 550 mg/m2). The elimination half-life of ENMD- 1198 averaged more than 12 hours after a single dose. Conclusions: DLT was identified at 550 mg/m2/d. Cohort expansion at 425mg/m2/d is ongoing. [Table: see text]


2018 ◽  
Vol 36 (4_suppl) ◽  
pp. 438-438 ◽  
Author(s):  
J. Randolph Hecht ◽  
Miklos Pless ◽  
Antonio Cubillo ◽  
Aitana Calvo ◽  
Steven Raman ◽  
...  

438 Background: T-VEC is a genetically modified HSV-1 oncolytic immunotherapy designed to preferentially replicate in tumors, produce GM-CSF, and stimulate anti-tumor immune responses. This study evaluates the safety of intrahepatic injection (inj) of T-VEC in patients (pts) with hepatocellular carcinoma (HCC) or liver metastases (mets). Methods: The primary objective is to assess the maximum tolerated dose. Eligible pts were ≥ 18 years (y) old, had progressive HCC or breast cancer (BC), colorectal cancer (CRC), gastroesophageal cancer, melanoma, non-small cell lung cancer, or renal cell cancer with liver mets, with measurable liver tumors suitable for inj. This dose escalation study comprised 2 groups: A (non-HCC) and B (HCC). T-VEC was given initially at 106 plaque-forming units (PFU)/mL followed by up to 4 mL of 107 PFU/mL (cohort 1) or 108 PFU/mL (cohort 2) every 21 (±3) days (Q21D), or up to 8 mL of the maximum tolerated concentration (MTC) Q21D (cohort 3). Inj volume was based on lesion size. Results: Results from cohorts 1 and 2 of group A are reported. 14 pts were treated; 12 (3 BC, 9 CRC) were DLT-evaluable: Median age was 65.5 y (range: 33, 73); median number of inj was 3; 1 pt received all 12 inj. MTC was 108 PFU/mL. There was 1 DLT, grade 3 aspartate aminotransferase (AST)/grade 2 bilirubin increase (inc), after 1 dose. In all treated pts, 4 (28.6%) had grade 3/4 treatment-related adverse events (TRAEs): anemia and inc gamma-glutamyltransferase, alanine aminotransferase (ALT), and AST. There were 2 deaths attributable to disease. Incidence of serious AEs (SAEs) is shown (Table). Conclusions: The MTC was 108 PFU/mL Q21D after initial inj at 106 PFU/mL. Repeated intrahepatic inj of T-VEC at the FDA-approved concentration for intralesional inj of melanoma was deemed tolerable and feasible in pts with liver mets. Additional investigation in combination with a PD-1 inhibitor is planned. Clinical trial information: NCT02509507. [Table: see text]


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4763-4763
Author(s):  
Rajshekhar Chakraborty ◽  
Ying Yan ◽  
Mike Royal

Abstract Background: With the advent of several new agents in the treatment of multiple myeloma, including proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies (mAb), the overall survival (OS) has significantly improved in the past two decades. However, most patients become refractory to currently available therapies along the disease trajectory. The median OS in triple-class-refractory patients (i.e., anti-CD 38 mAb, PIs, and IMiDs) is 6 months (Gandhi et al. Leukemia. 2019). Although BCMA-targeted therapies are a major advance in such patients, none of them are curative thus far, with the median progression-free survival (PFS) ranging from 3 months in BCMA-antibody drug conjugates (ADCs) to approximately 1 year in BCMA chimeric antigen receptor T-cell (CAR T-cell) therapies. Furthermore, unique toxicities of BCMA-targeted agents such as ocular toxicity (ADCs), cytokine release syndrome, or neurotoxicity (CAR Ts and BiTEs) may preclude their use in many patients. Hence, there is a critical unmet need in patients with triple-class-refractory myeloma. The STI-6129 ADC is produced by conjugating STI-5171, a fully human anti-CD38 mAb, with a covalently bound tubulin inhibitor, duostatin 5.2 (DUO-5.2), using a proprietary linker technology with a 3:1 drug-antibody ratio. STI-6129 binds to different CD38 epitopes than daratumumab. Upon binding to CD38, STI-6129 ADC is internalized by the cancer cell and undergoes lysosomal degradation, releasing DUO-5.2. This in turn leads to G2-phase cell cycle arrest, followed by caspase 3/7-dependent apoptosis and cell death. In studies on cynomolgus monkeys, the serum level of DUO-5.2 remained undetectable at all doses except at the highest dose of 4.5 mg/kg, indicating low likelihood of off-target toxicity. In vitro studies of primary human plasma cells, human tumor models, and animal xenograft models have demonstrated target elimination of CD38-positive human plasma cells by STI-6129. Importantly, STI-6129 has demonstrated cytotoxic activity in human multiple myeloma cells isolated from daratumumab-refractory patients (Figure 1). Hence, further investigation on clinical activity of STI-6129 is warranted. Study Design: The 38-ADC-RRMM-101 study is a two-stage, multicenter, open-label, dose-finding phase 1/2 trial. The phase 1 trial is designed to identify the recommended phase 2 dose (RP2D) of STI-6129 by assessing safety, preliminary efficacy, and pharmacokinetics in patients with relapsed/refractory (RR) myeloma. The phase 2a stage of the trial will be a single-arm study to investigate the efficacy of STI-6129 in an expansion cohort of RR myeloma patients. Up to 25 patients will be enrolled in the phase 1 stage and 30 in the phase 2 stage of the study. The key inclusion criteria are the following: (a) RR myeloma with at least 3 prior lines of therapy in addition to being refractory to at least 1 PI, 1 IMiD, and 1 anti-CD 38 mAb; (b) measurable disease. The key exclusion criteria are: (a) receipt of the last dose of any anti-CD mAb within 90 days; (b) grade ≥3 neuropathy or grade 2 neuropathy with pain; (c) current history of CTCAE grade 3 muscle paresis, eyelid conditions, glaucoma, or any other ocular disorder that is CTCAE grade 2; (d) estimated creatinine clearance &lt;60 ml/min; (e) left ventricular ejection fraction&lt;40%. The primary endpoint of phase 1 stage of the study is safety, particularly any dose limiting toxicities. The primary endpoint of the phase 2 part of the study is overall response rate as per IMWG criteria. The dosing cohorts in the dose escalation phase will range from 0.25 mg/kg to 3.68 mg/kg. Given the potential for on-target off-tumor toxicity due to expression of CD38 on non-clonal plasma cells and other hematopoietic cells, blood counts and immunoglobulin levels will be closely monitored. While neurotoxicity or ocular toxicity were not observed with STI-6129 in animal models, including non-human primates, such event will be considered as adverse events of special interest (AESI) and comprehensive neurology and ocular (including slit lamp) examinations will be performed at baseline and study completion, and at any AESI. STI-6129 will be administered intravenously once in a 4-week cycle, with the intention being to treat patients until disease progression or unacceptable toxicity. Figure 1 Figure 1. Disclosures Yan: Sorrento Therapeutics: Current Employment. Royal: Sorrento Therapeutics: Current Employment.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e13604-e13604 ◽  
Author(s):  
Muralidhar Beeram ◽  
Amita Patnaik ◽  
Ravi K. Amaravadi ◽  
Naomi B. Haas ◽  
Kyriakos P. Papadopoulos ◽  
...  

e13604 Background: MGCD265 is a multi-target oral tyrosine kinase receptor inhibitor that targets Met, VEGFRs 1, 2, 3, Tie and Ron. The maximum tolerated dose (MTD) of MGCD265 in combination with docetaxel was initially determined using micronized tablets. MGCD265 formulation was improved and the MTD of MGCD265 in combination with docetaxel using the updated formulation was re-evaluated. Methods: Patients (pts) with metastatic or advanced solid tumors were enrolled in this Phase I, open-label, dose-escalating study to assess safety, pharmacodynamics (PD) and pharmacokinetics (PK) as well as anti-tumor activity of the combination. MGCD265 was administered every day over a 3-week cycle and docetaxel (50 then 75 mg/m2) was given intravenously once every 3 weeks (q3w). Results: As of January 11, 2012, 34 pts were enrolled (M/F: 19/15; ECOG 0/1: 19/15; median age: 64 years old). The MTD of the combination was initially defined as MGCD265 (72 mg/m2 BID)+docetaxel (75 mg/m2 q3w) based on the occurrence of dose limiting toxicities (DLTs) in 2 pts who were treated with MGCD265 at 96 mg/m2 BID (fatigue in 1 pt and diarrhea & lipase elevation in the other pt). The updated formulation of MGCD265 was introduced at 48 mg/m2 BID (n=3) with dose escalation to 72 mg/m2 BID (n=3) and 96 mg/m2 (n=4) with no observed DLTs, though the exposure of the two formulations were generally comparable. Overall, objective partial responses (per RECIST 1.1) were observed in 2/9 pts with NSCLC, 1/3 pts with prostate cancer and 1/1 pt with endometrial cancer. Stable disease for 6 cycles or more was observed in 6 pts. Treatment-related ≥ grade 3 toxicity reported in ≥ 2 pts were neutropenia, leukopenia, diarrhea and elevated lipase. The PD profile indicated an increase in the plasma level of VEGF and a decrease in plasma level of HGF at Cycle 1 Day 8 compared to baseline in some patients. Conclusions: MGCD265 was found to be well tolerated using the updated formulation in combination with full-dose docetaxel. Anti-cancer activity, supporting Phase II development of the combination, was observed.


Blood ◽  
2012 ◽  
Vol 120 (3) ◽  
pp. 552-559 ◽  
Author(s):  
Jeffrey A. Zonder ◽  
Ann F. Mohrbacher ◽  
Seema Singhal ◽  
Frits van Rhee ◽  
William I. Bensinger ◽  
...  

Abstract This multicenter, first-in-human study evaluated the safety, tolerability, and pharmacokinetic and pharmacodynamic properties of the anti-CS1 monoclonal antibody elotuzumab. A standard 3 + 3 design was used to determine maximum tolerated dose; dose-limiting toxicities were assessed during cycle 1. Thirty-five patients with relapsed/refractory multiple myeloma were treated with intravenous elotuzumab at doses ranging from 0.5 to 20 mg/kg every 2 weeks. Patients who achieved at least stable disease after 4 treatments could receive another 4 treatments. No maximum tolerated dose was identified up to the maximum planned dose of 20 mg/kg. The most common adverse events, regardless of attribution, were cough, headache, back pain, fever, and chills. Adverse events were generally mild to moderate in severity, and adverse events attributed to study medication were primarily infusion-related. Plasma elotuzumab levels and terminal half-life increased with dose whereas clearance decreased, suggesting target-mediated clearance. CS1 on bone marrow–derived plasma cells was reliably saturated (≥ 95%) at the 10-mg/kg and 20-mg/kg dose levels. Using the European Group for Bone and Marrow Transplantation myeloma response criteria, 9 patients (26.5%) had stable disease. In summary, elotuzumab was generally well tolerated in this population, justifying further exploration of this agent in combination regimens.


Sign in / Sign up

Export Citation Format

Share Document