SAKK 65/08: A Phase I Trial of the HIV Protease Inhibitor Nelfinavir in Combination with Bortezomib Identifies Nelfinavir As FDA Approved, Oral Drug that Inhibits the Proteasome and Induces Proteotoxic Stress in Vivo and has Potential Antimyeloma Activity.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2956-2956
Author(s):  
Christoph Driessen ◽  
Dagmar Hess ◽  
Thomas Pabst ◽  
Sarah R Haile ◽  
Markus Joerger ◽  
...  

Abstract Abstract 2956 Introduction: The HIV protease inhibitor nelfinavir has anti-myeloma activity in mice; it is approved at the 1250 mg bid dose for oral treatment of HIV. We performed a phase I dose escalation trial of nelfinavir in combination with bortezomib in patients with advanced hematologic malignancies. Methods: During cycle 1 (28 days), trial treatment consisted of 1 week nelfinavir monotherapy, followed by nelfinavir in combination with standard dose bortezomib (1.3 mg/m2i.v. day 8, 11, 15, 18), while cycles 2 and 3 (21 days each) consisted of 2 weeks nelfinavir in combination with bortezomib (day 1, 4, 8, 11). Non-progressing patients could continue therapy for up to 4 additional cycles with the same regimen as cycles 2 and 3. Nelfinavir dose was escalated in a 3+3 design over 3 dose levels (1250, 1875, 2500 mg bid). Dose limiting toxicity (DLT), the primary endpoint, was grade 3–4 non-hematological toxicity (excluding grade 3 bilirubin/alanine aminotransferase (ALT) or hyperlipidemia reversible within 2 weeks) or severe hematologic toxicity unrelated to the underlying disease during cycle 1. Secondary endpoints included pharmacodynamic and pharmacokinetic assessments during cycle 1 at baseline, nelfinavir monotherapy and after application of nelfinavir and bortezomib in combination, as well as signals for activity. Results: Twelve evaluable patients were registered (median age 58 years; 8 male; performance status 0–1 in 10/12 patients); 8 had multiple myeloma, 2 leukemia (1 acute myeloid, 1 acute lymphoblastic) and 2 lymphoma (1 diffuse large B-cell lymphoma, 1 mantle-cell (MCL)). All myeloma patients failed both prior bortezomib and lenalidomide-containing therapy; 7/8 had progressed under prior bortezomib. One patient (2500 mg bid dose) experienced a transient grade 4 elevated ALT, categorized as DLT, which resolved within 2 weeks. The patient continued the same regimen off study without recurrent hepatic toxicity. No further DLTs occurred, thus nelfinavir 2500 mg bid was established to be safe in combination with standard dose bortezomib. One patient with highly aggressive lymphoma died from cerebral vein thrombosis; a myeloma patient experienced a non-fatal pulmonary embolism. Elevated ALT (2 patients) was the only additional non-hematological toxicity grade 3/4 observed in >1 patient. Grade 3 febrile neutropenia and grade 4 thrombocytopenia were seen in 1 and 4 patients, respectively. Best treatment response was evaluated for 11 patients (1 not evaluable). Partial response was achieved in 3 patients (2 myeloma, 1 MCL) and stable disease for at least 2 cycles of therapy in 5 patients. Overall, 4/12 patients completed >=3 cycles of treatment. Assessment of proteasome activity in peripheral blood mononuclear cells (PBMC) from treated patients after 1 week nelfinavir monotherapy revealed inhibition of total proteasome activity in vivo by nelfinavir compared to baseline (mean inhibition, as determined by specific, quantitative intracellular affinity labeling of active proteasome subunits: 14.9 %, 95% confidence interval (CI): 8.8–23.5%, p=<0.001), including inhibition of the bortezomib-insensitive tryptic (β2-type) proteasome activity (mean inhibition: 17.7%, 95% CI: 8.0–27.4%, p=0.008). In addition, inhibition of pAKT, induction of the unfolded protein response and accumulation of polyubiquitinated protein in vivo was observed in PBMC after nelfinavir monotherapy. Mean intracellular proteasome inhibition after combination treatment with bortezomib and nelfinavir was 26.6 % (95% CI: 11.5–42%). Maximum nelfinavir plasma levels were observed at the 1875 mg bid dose level (Cmax mean 12.10 mM, trough mean 6.97 mM), matching the nelfinavir concentrations that mediate anti-myeloma activity in vitro. Conclusion: This is the first trial to report on the use of nelfinavir as an anti-neoplastic agent in patients with hematologic malignancies. It identifies nelfinavir as FDA approved, orally available drug with pan-proteasome inhibiting activity in vivo. Nelfinavir treament up to 2500 mg bid is safe as monotherapy and in combination with standard dose bortezomib. Bortezomib in combination with nelfinavir shows signals for clinical activity in individual myeloma patients that have failed bortezomib and lenalidomide-containing therapies. Nelfinavir warrants further clinical investigation in multiple myeloma, in particular in combination with proteasome inhibitors. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2898-2898 ◽  
Author(s):  
Swaminathan Padmanabhan ◽  
Kevin R Kelly ◽  
Mark Heaney ◽  
Stephanie Hodges ◽  
Suzanne Chanel ◽  
...  

Abstract Abstract 2898 Background: STA-9090 is a potent, second-generation, small-molecule Hsp90 inhibitor, with a chemical structure unrelated to the first-generation, ansamycin family of Hsp90 inhibitors (e.g., 17-AAG or IPI-504). STA-9090 induces the loss of Hsp90 client proteins that are important in hematologic cancers, including BCR-ABL, c-KIT, FLT3, WT1, and JAK2. In preclinical studies, STA-9090 has shown potency up to 100 times greater than the first-generation Hsp90 inhibitors as well as activity against a wider range of kinases. In in vitro and in vivo models, STA-9090 has shown potent activity against a broad range of leukemias, lymphomas, and multiple myeloma. Methods: The primary objective of this Phase I multicenter study (NCT00858572) was to determine the maximum tolerated dose (MTD) of STA-9090 given twice weekly in patients with acute myeloid leukemia (AML), advanced myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML), and myeloproliferative neoplasms (MPN). An adaptive statistical design methodology was used to determine dose escalation. Safety, pharmacokinetics (PK), pharmacodynamic (PD), and clinical activity were evaluated. Dosing for 4 consecutive weeks constituted one dosing cycle. Serial pharmacokinetic (PK) samples were collected pre-dose, 0.5, 1, 1.5, 2, 4, 6, 8, and 24 hours post-dose on study days 1 and 22, and pre-dose and 1 hour post at all other visits in cycle 1. Plasma samples were collected for HSP70 protein analysis. Safety assessments included the number and grade of adverse events (AEs), changes from baseline in laboratory parameters, and evaluation of electrocardiogram changes. Results: 22 patients (14 males, 8 females; median age 62 years, range 33–80; Eastern Cooperative Oncology Group [ECOG] status range [0-2]) received STA-9090 twice weekly. Patients with the following disease types were treated: AML (n=7), CML (n=8), MDS (n=3), and MPN (n=4). The median time from initial diagnosis to first treatment was 19 months; patients had received a median of 2 (range, 1–9) prior treatments and 27% were refractory to their most recent therapy. Dosing with STA-9090 was at 14 mg/m2 (4 patients), 22 mg/m2 (4 patients), 30 mg/m2 (3 patients), 70mg/m2 (3 patients), and 110 mg/m2 (8 patients). Patients received a median of 2 (range, 1–8) cycles of STA-9090. AEs reported in ≥25% of patients were diarrhea, nausea, fatigue, abdominal pain, anemia, increased alanine aminotransferase, arthralgia, increased aspartate aminotransferase, dyspnea, hypokalaemia, thrombocytopenia, and vomiting. The majority of AEs were mild to moderate in severity. There have been 3 patients with dose limiting toxicities: one at 14 mg/m2 (grade 3 hyponatremia and hyperbilirubinemia) and two at 110 mg/m2 (grade 3 prolonged QTc and transaminitis). At the time of abstract submission, the MTD has not been defined. STA-9090 exhibited linear PK with exposures (AUC and Cmax) increasing in proportion to dose. One hour following infusion termination, plasma concentrations declined by approximately 10 fold, and by 100 fold within approximately 8 to 10 hours. STA-9090 showed no drug accumulation with twice weekly dosing. STA-9090 was well distributed, exhibiting an apparent volume of distribution greater than total body water. HSP70 protein levels increased following STA-9090 administration and remained elevated prior to subsequent doses indicating a durable biological impact of the drug. Clinical responses include 1 patient with CML (dosed at 14 mg/m2) and 1 patient with AML (dosed at 110 mg/m2) who had best responses of hematological improvement lasting 2 and 3 months, respectively. In addition, 4 patients with myelofibrosis (1 patient dosed at 14 mg/m2 and 3 dosed at 70 mg/m2) had stable disease as best response; 1 of these responses lasted for 7 months and the patient went on to receive an allogenic stem cell transplant. Further response evaluation is ongoing. Conclusions: In patients with hematologic malignancies, STA-9090 is well tolerated up to dose levels of 110 mg/m2 given twice weekly. Induction of HSP70 protein levels provides evidence of in vivo biological activity. A favorable PK profile, preliminary signs of pharmacodynamic activity, and early clinical activity signals warrant continued evaluation of single-agent STA-9090 using a twice weekly dosing regimen. Disclosures: Off Label Use: Phase I clinical study. Bradley:Synta Pharmaceuticals Corp.,: Employment, Equity Ownership. Teofilovici:Synta Pharmaceuticals Corp.,: Employment, Equity Ownership.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1971-1971
Author(s):  
Wolfram Pönisch ◽  
Simone Heyn ◽  
Ina Wagner ◽  
Martin Mohren ◽  
Franz-Albert Hoffmann ◽  
...  

Abstract Abstract 1971 Introduction: While the role of lenalidomide monotherapy in the treatment of relapsed/refractory patients with multiple myeloma (MM) is established, combination therapies with Lenalinomide are still under investigation. Bendamustine is a bi-functional alkylating agent with a purine-like benzimidazole ring effective in combination with steroids, thalidomide and bortezomib for the treatment of patients with MM. In the current trial, combination therapy of bendamustine, lenalinomide and prednisolone (RBP) was tested for feasibility and safety in patients with relapsed or refractory MM. Patients and Methods: This is a phase I trial examining dosing of lenalidomide in combination with bendamustine and prednisolone. The first cohort of patients received a starting dose of 10mg/d d1-21 lenalidomide, 60mg/m2/d d1-2 bendamustine and 100mg/d d1-4 prednisolone. Escalation steps in the next cohorts included 15, 20 and 25mg of lenalidomide followed by an escalation step of 75 mg/m2 bendamustine. Three patients were enrolled at each dose level and the first two cycles were evaluated for maximum tolerable dose. Patients received RBP in 4-week cycles for a maximum of 8 cycles in order to evaluate efficacy. Patients with stable or responding disease following 8 cycles of RBP received single-agent oral lenalidomide 10 mg once daily on days 1–21 of each 28-day cycle as maintenance. Results: : Nine patients (3 at each dose level of 10 mg, 15 mg or 20 mg lenalidomide) have been enrolled to date and 9 patients have completed at least 2 cycles. Response was assessed using modified EBMT criteria to include near complete remission (nCR) and very good partial remission (VGPR). 8 of 9 patients responded after at least 2 cycles with 2 VGPR, 4 PR, 1 MR and 1 stable disease. One patient experienced progressive disease. None of the 9 patients developed dose-limiting hematoxicity as defined by an ANC < 1,0 × 109/l with fever for > 3 days or an ANC <0,5 × 109/l for > 7 days or platelet count < 25 × 109/l for > 3 days. Neutropenia was reported in 4 patients (CTC grade ≥ 3) but no thrombocytopenia (CTC grade ≥ 3) was observed. No grade 3 or 4 non hematological toxicity was encountered and no dose modification was required. Conclusions: RBP with a dose of 20 mg lenalidomide d 1–21 and 60 mg/m2 bendamustine d 1–2 is well tolerated in patients with relapsed or refractory MM. Maximum tolerable dose was not reached. Further dose increase according to the protocol is in progress. Disclosures: Niederwieser: Bristol-Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 2018-2018 ◽  
Author(s):  
K. Kato ◽  
T. Hamaguchi ◽  
H. Yasui ◽  
T. Okusaka ◽  
H. Ueno ◽  
...  

2018 Background: NK105 is a new polymeric micelle carrier system for paclitaxel (PTX). A preclinical study revealed that the plasma AUC and tumor AUC of NK105 were 90-fold higher and 25-fold higher, respectively, than those of free-PTX, i.e., the conventional PTX formulation. NK105 had higher in vivo antitumor activity and lower neurotoxicity than free-PTX. This phase I study was designed to examine the MTD, DLTs, recommended dose (RD) for phase II, and the pharmacokinetics of NK105. Methods: NK105 was administered as a 1-hour intravenous infusion every 3 weeks, without antiallergic premedication. The starting dose was 10 mg PTX equivalent/m2, and the dose was escalated according to the accelerated titration method. Results: To date, 17 patients (pts) have received the following doses: 10 mg/m2 (n=1); 20 mg/m2 (n=1); 40 mg/m2 (n=1); 80 mg/m2 (n=1); 110 mg/m2 (n=3); 150 mg/m2 (n=5); and 180 mg/m2 (n=5). The tumor types treated included pancreatic (n=9), bile duct (n=5), gastric (n=2), and colonic (n=1) cancers. Neutropenia was the most common hematological toxicity. Grade 3 fever developed in 1 pt given 180 mg/m2. No other grade 3 or 4 non-hematological toxicity, including neuropathy, was observed. DLTs occurred in pts given 180 mg/m2 (grade 4 neutropenia lasting for more than 5 days). This dose was designated as the MTD. Allergic reactions developed in only one pt at 180 mg/m2, who was sensitive to other drugs such as antibiotics anti-inflammatory. A partial response was observed in one pt with pancreatic cancer and pts with colonic and gastric cancer had stable disease. The Cmax and AUC of NK105 were dose dependent. The plasma AUC of NK105 at 180 mg/m2 was approximately 30-fold higher than that of the conventional formulation of PTX. Conclusions: Accrual is ongoing at the 150 mg/m2 dose level to determine the RD. DLT was Grade 4 neutropenia. NK105 produces prolonged high levels of PTX in plasma. A 1-hour infusion of NK105 every 3 weeks was feasible, well tolerated, and effective in patients with pancreatic cancer. Even after the long term usage, only grade 1 or 2 neuropathy was observed. NK105 will be evaluated in Phase II studies of patients with advanced pancreatic and gastric cancers. No significant financial relationships to disclose.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3019-3019 ◽  
Author(s):  
Massimo Offidani ◽  
Federica Cavallo ◽  
Claudia Polloni ◽  
Anna Marina Liberati ◽  
Stelvio Ballanti ◽  
...  

Abstract Abstract 3019 Background: Panobinostat (LBH-589) is a pan-deacetilase inhibitor that targets histone proteins increasing tumor suppressor gene activities leading to cell-cycle and differentiation arrest besides to target non-histone proteins such as HSP90, aggressomes, p53, HIF-1a, and a-tubulin somehow promoting cell death. Panobinostat, combined with steroids and/or immunomodulatory drugs, demonstrated additive/synergistic activity in Multiple Myeloma (MM) and ability to overcome previous chemoresistance. Several combination studies with Panobinostat plus novel drugs are now ongoing in MM. Methods: This is a multicenter, open-label, phase I-II study exploring the combination of a standard therapy such as MPT (Melphalan 0.18 mg/kg per os for 4 days, Prednisone 1.5 mg/kg per os for 4 days, Thalidomide 50 mg/day continuously) with Panobinostat 15 mg p.o. thrice weekly for 3 weeks in a 28-day cycle to assess safety profile and activity of this combination in patients with relapsed/refractory MM having adequate performance status and haematological, cardiac, liver and neurological functions. The study was designed according to the Briant and Day method that plans a “dose-escalation phase” to determine both the MTD and the activity of the study drug and an “expansion-phase” in which the MTD of the study drug is used to further assess its safety and efficacy. Despite in the first phase of this study 19 patients were planned according to the study design, protocol was amended after 13 patients had been enrolled since more than 50% grade 3–4 toxicity occurred although response criteria were met. Therefore, Panobinostat was reduced to 10 mg p.o. thrice weekly for 3 weeks in a 28-day cycle whereas the dose of drugs of the MPT combination was not modified. Toxicity and response were assessed according to CTC version 4 and IMWG criteria, respectively. Results: As of February 2010, 24 patients were enrolled in this study. Median age was 71.5 years (range 40–81 years) and 12 patients (50%) had ISS 2–3 score. Patients had received a median of 2 prior therapies (range 1–6) and 5 (21%) three or more prior lines of therapy. Sixteen (73%), 13 (54%), 18 (75%), 11 (46%) and 9 (37.5%) patients had been previously treated with ASCT, thalidomide, bortezomib, lenalidomide and all 3 new-drugs, respectively. Seven patients (29%) were refractory to the last therapy. Twelve patients (50%) had a disease history longer than 5 years. In the first 13 patients treated with Panobinostat 15 mg, grade 3–4 thrombocytopenia and neutropenia occurred in 6 (46%) and 9 patients (69%), respectively. Moreover, 4 patients (31%) developed non-hematological adverse events such as fatigue, constipation, infection and arrythmia. In the group of 11 patients treated with Panobinostat 10 mg, grade 3–4 thrombocytopenia decreased to 18% (2 patients) but neutropenia was still high (8 patients: 72.5%). Three patients (27%) had grade 3–4 non-hematological toxicity (one fatigue and two constipation). No patients had QTcF prolongation or severe neuropathy. Dose adjustment was necessary in 9 patients (37.5%, all due to hematological toxicity) while 6 patients (25%) interrupted the protocol because of side effects (5 due to no resolution of grade 3–4 hematological toxicity within 4 weeks and one due to atrial fibrillation). One patient (4%) died on study due to sepsis during prolonged neutropenia. Response ≥ PR were observed in 12 patients (50%) including 4 VGPR and 8 PR. Additionally, 2 patients had MR and 8 SD. Only 2 patients progressed during treatment. There was no difference between the two cohorts of patients (Panobinostat 15 mg and Panobinostat 10 mg) in terms of response ≥ PR (54% vs 45.5%) or disease progression (7.5% vs 9%). Notably, response was obtained also in 2/7 patients (28%) who progressed during bortezomib or IMIDs. Conclusions: This study suggests that MPT-Panobinostat combination has an encouraging anti-myeloma activity since responses were still seen in patients with advanced stage or resistant to new drugs diseases. Different schedules of Panobinostat/melphalan should be explored to reduce haematological toxicity. Disclosures: Offidani: Celgene: Honoraria. Off Label Use: Panobinostat in relapsed/refractory multiple myeloma. Cavallo:Celgene: Honoraria. Polloni:Celgene: Honoraria. Ballanti:Celgene: Honoraria. Catarini:Celgene: Honoraria. Alesiani:Celgene: Honoraria. Corvatta:Celgene: Honoraria. Gentili:Celgene: Honoraria. Boccadoro:Celgene: Honoraria, Research Funding. Leoni:Celgene: Honoraria. Palumbo:Celgene: Honoraria, Research Funding.


AIDS ◽  
1996 ◽  
Vol 10 (5) ◽  
pp. 485-492 ◽  
Author(s):  
Daniel S. Stein ◽  
Douglas G. Fish ◽  
John A. Bilello ◽  
Sandra L. Preston ◽  
Gisele L. Martineau ◽  
...  

1998 ◽  
Vol 42 (12) ◽  
pp. 3218-3224 ◽  
Author(s):  
Hing L. Sham ◽  
Dale J. Kempf ◽  
Akhteruzammen Molla ◽  
Kennan C. Marsh ◽  
Gondi N. Kumar ◽  
...  

ABSTRACT The valine at position 82 (Val 82) in the active site of the human immunodeficiency virus (HIV) protease mutates in response to therapy with the protease inhibitor ritonavir. By using the X-ray crystal structure of the complex of HIV protease and ritonavir, the potent protease inhibitor ABT-378, which has a diminished interaction with Val 82, was designed. ABT-378 potently inhibited wild-type and mutant HIV protease (Ki = 1.3 to 3.6 pM), blocked the replication of laboratory and clinical strains of HIV type 1 (50% effective concentration [EC50], 0.006 to 0.017 μM), and maintained high potency against mutant HIV selected by ritonavir in vivo (EC50, ≤0.06 μM). The metabolism of ABT-378 was strongly inhibited by ritonavir in vitro. Consequently, following concomitant oral administration of ABT-378 and ritonavir, the concentrations of ABT-378 in rat, dog, and monkey plasma exceeded the in vitro antiviral EC50 in the presence of human serum by >50-fold after 8 h. In healthy human volunteers, coadministration of a single 400-mg dose of ABT-378 with 50 mg of ritonavir enhanced the area under the concentration curve of ABT-378 in plasma by 77-fold over that observed after dosing with ABT-378 alone, and mean concentrations of ABT-378 exceeded the EC50 for >24 h. These results demonstrate the potential utility of ABT-378 as a therapeutic intervention against AIDS.


Sign in / Sign up

Export Citation Format

Share Document