2006 RADAR Report Card on Conversion Rates from Accelerated FDA Approval to Regular FDA Approval for Drugs Used To Treat Hematologic Malignancies: Policy Changes Continue To Be Needed.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3318-3318
Author(s):  
Andrew J. Lurie ◽  
Mark V. Mai ◽  
Martin S. Tallman ◽  
June McCoy ◽  
Charles L. Bennett

Abstract Background: At ASH 2004 and 2005, we reported < 20% rates of from accelerated approval (AA) to regular approval (RA) for drugs used to treat hematologic malignancies (HMs). The FDA convened Oncologic Drug Advisory Committee (ODAC) meetings with sponsors of these drugs in 2003 and 2005 outlining recommendations for completion of subpart H trials. Congressmen Markey and Hinchey have raised concern over low rates of completion of subpart H studies for all AA drugs. The Research on Adverse Drug Events and Reports (RADAR) project reviewed the current status of subpart H commitments for all cancer drugs that received AA prior to 2005. Methods: NDAs and supplements for new uses from the FDA, package inserts, and literature reviews were reviewed. Results: Of 19 cancer drugs receiving AA prior to 2005, conversion to RA has occurred for 2 of 9 drugs for HMs and 8 of 11 drugs for solid tumors (STs) (22% vs 73%, p<0.05). Subpart H studies have been ongoing for a median of 5 years for AA drugs for HMs; however, for ST drugs, these studies were completed in a median of 2.5 years. Of 7 subpart H studies reviewed at the 2005 ODAC meeting on AA, subpart H evaluations continue for all 4 for HMs versus 1 of 3 for STs. HMs are characterized by lower rates of target populations, most having > 25,000 newly diagnosed patients annually (25% vs 78%). Recruitment targets are smaller for AA studies for HMs (median, 82 vs 295 patients) and subpart H studies (median, 295 vs 535). Conclusions: Rates of conversion from AA to RA remain poor for drugs for hematologic malignancies but not for solid tumors drugs. In spite of ODAC meetings and two congressional inquiries, little progress ahs been made towards completing these trials. RADAR continues to urge policy makers to modify the AA process for drugs used to treat hematologic malignancies. Hematologic malignancy drugs receiving AA prior to 2005 and current status of subpart studies Drug AA Indication Pts in AA trial design AA year Target # of pts in Subpart H study RA Year Imatinib mesylate CML 1027 2001 1027 2003 Bortezomib MM 202 2003 669 2005 Denileukin difitox* CTCL** 71 1999 195 Pending Liposomal cytarabine* Lymphomatous meningitis** 33 1999 195 Pending Gemtuzumab ozogamicin* AML** 142 2000 342 Pending Alemtuzumab* B-Cell Chronic lymphocytic leukemia** 93 2001 197 Pending Ibritumomab tiuxetan Certain NHLs ** 230 2002 293 Pending Imatinib mesylate CML (pedatrics)** 39 2003 35 Pending Tositumomab Certain NHLs** 60 2003 N/A Pending Solid tumor cancer drugs receiving AA prior to 2005 and current status of subpart studies Drug AA Indication Pts in AA trial design AA year Target # of pts in Subpart H study RA year * Recommendations for completing trials were made at recent ODAC meetings ** Indications with >25,000 new diagnosis annually Irinotecan Colorectal 304 1996 535 1998 Docetaxel Breast 134 1996 326 1998 Capecitabine Breast 163 1998 511 2001 Temozolomide Brain 162 1999 573 2005 Liposomal Doxorubicin* Ovarian 85 1999 474 2005 Oxaliplatin Colorectal 463 2002 795 2004 Anastrozole Breast 9366 2002 6196 2005 Gefitinib Lung 221 2003 1700 2005 Liposomal Doxorubicin* Kaposi’s sarcoma** 77 1995 250 Pending Amifostine Non-small cell lung** 25 1996 366 Withdrawn Imatinib mesylate GI stromal tumors** 147 2002 946 Pending

2018 ◽  
Vol 36 (18) ◽  
pp. 1805-1812 ◽  
Author(s):  
Thomas J. Hwang ◽  
Jessica M. Franklin ◽  
Christopher T. Chen ◽  
Julie C. Lauffenburger ◽  
Bishal Gyawali ◽  
...  

Purpose The breakthrough therapy program was established in 2012 to expedite the development and review of new medicines. We evaluated the times to approval, efficacy, and safety of breakthrough-designated versus non–breakthrough-designated cancer drugs approved by the US Food and Drug Administration (FDA). Methods We studied all new cancer drugs approved by the FDA between January 2012 and December 2017. Regulatory and therapeutic characteristics (time to FDA approval, pivotal trial efficacy end point, novelty of mechanism of action) were compared between breakthrough-designated and non–breakthrough-designated cancer drugs. Random-effects meta-regression was used to assess the association between breakthrough therapy designation and hazard ratios for progression-free survival (PFS), response rates (RRs) for solid tumors, serious adverse events, and deaths not attributed to disease progression. Results Between 2012 and 2017, the FDA approved 58 new cancer drugs, 25 (43%) of which received breakthrough therapy designation. The median time to first FDA approval was 5.2 years for breakthrough-designated drugs versus 7.1 years for non–breakthrough-designated drugs (difference, 1.9 years; P = .01). There were no statistically significant differences between breakthrough-designated and non–breakthrough-designated drugs in median PFS gains (8.6 v 4.0 months; P = .11), hazard ratios for PFS (0.43 v 0.51; P = .28), or RRs for solid tumors (37% v 39%; P = .74). Breakthrough therapy–designated drugs were not more likely to act via a novel mechanism of action (36% v 39%; P = 1.00). Rates of deaths (6% v 4%; P = .99) and serious adverse events (38% v 36%; P = 0.93) were also similar in breakthrough-designated and non–breakthrough-designated drugs. Conclusion Breakthrough-designated cancer drugs were associated with faster times to approval, but there was no evidence that these drugs provide improvements in safety or novelty; nor was there a statistically significant efficacy advantage when compared with non–breakthrough-designated drugs.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3897-3897
Author(s):  
David H. Henry ◽  
Susan Kilcoyne ◽  
Jonathan N. Latham ◽  
Arthur P. Staddon

Abstract Pegylated liposomal doxorubicin (PLD, Doxil) was developed to decrease the risk of cumulative-dose cardiotoxicity associated with conventional doxorubicin. However, hand-foot syndrome (HFS) and stomatitis were associated with PLD in early clinical trials and were more common when patients with ovarian cancer received 50 mg/m2 q4w (HFS, 37.4%; stomatitis, 37.4%) than when patients with AIDS-related Kaposi’s sarcoma (KS) received 20 mg/m2 q3w (HFS, 3.4%; stomatitis, 6.8%). PLD is now commonly used with a dose intensity of 10 mg/m2 per week (20 mg/m2 q2w, 30 mg/m2 q3w, or 40 mg/m2 q4w). Recent reports with this reduced dose intensity have shown a marked decrease in the rate and grade of HFS and stomatitis. This retrospective chart review was performed to examine the tolerability and effectiveness of PLD 20 mg/m2 q2w in patients with KS and hematologic malignancies. Patient charts from a community oncology clinic from January 2000 through December 2004 were reviewed. Data abstracted included patient demographics, PLD dosing, treatment response, and tolerability for all PLD recipients. Data were censored for patients with solid tumors or an initial PLD dose other than 20 mg/m2 q2w. Of the 157 patients receiving PLD, 65 received 1206 cycles (range, 1–116; median, 17) of PLD at an initial dose of 20 mg/m2 every 2 weeks as a component of care for KS (n=55), multiple myeloma (n=4), non-Hodgkin’s lymphoma (NHL, n=4), Hodgkin’s disease (n=1), or chronic lymphocytic leukemia (n=1). Most patients with KS also received highly active antiretroviral therapy (HAART). No signs (eg, decreased ejection fraction) or treatment-related symptoms (eg, shortness of breath) of cardiotoxicity were reported in any patient. Four patients (6.2%) had documented mild HFS; 3 patients with KS and 1 patient with a hematologic malignancy One case of HFS (1.5%) led to discontinuation of PLD. Three patients (4.6%) had documented symptoms of mucositis; all 3 patients had KS (5.5%). PLD was effective in most patients at this dose intensity; clinical response (complete or partial response) was seen in 55 patients (84.6%), stable disease in 1 patient (1.5%), and progression in 3 patients (4.6%, all with NHL), and response could not be determined in 5 patients (7.7%). The results of this retrospective review suggest that PLD at an initial dose of 20 mg/m2 q2w is active and well tolerated, and that HFS and stomatitis occur in a low percentage of patients and rarely result in discontinuation. Moreover, the rates of HFS seen in patients with KS and hematologic malignancies seem similar. Prospective studies in larger populations are required to confirm the efficacy and safety of PLD 20 mg/m2 q2w as a component of care for hematologic malignancies. Incidences of Targeted Adverse Events During Treatment with PLD 20 mg/m2 KS (n = 55) Hematologic Malignancies (n = 10) Cardiotoxicity 0 (0.0%) 0 (0.0%) Mucositis 3 (5.5%) 0 (0.0%) HFS 3 (5.5%) 1 (10.0%)


Author(s):  
Emre Yekedüz ◽  
Dario Trapani ◽  
Wenxin Xu ◽  
Elisabeth GE Vries ◽  
Chris Labaki ◽  
...  

2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 2580-2580
Author(s):  
B. Ramaswamy ◽  
M. Phelps ◽  
R. Baiocchi ◽  
T. Bekaii-Saab ◽  
D. Wilkins ◽  
...  

2580 Background: A phase I study of flavopiridol, a cyclin-dependent kinase inhibitor, using an alternative schedule was conducted in pts with solid tumors given its promising activity in pts with chronic lymphocytic leukemia (CLL). Methods: Using standard 3x3 ph I dose escalation design, NCI-sponsored trial was performed to determine the safety and dose-limiting toxicity (DLT) of flavopiridol given as a 30-min IV loading dose followed by a 4-hr infusion weekly for 4 wks repeated every 6 wks. DLT was defined as Gr 4 hematologic toxicity (HT) for > 7 days, > Gr 3 non-HT except Gr 3 fatigue or diarrhea resolving <4 days and cytokine release syndrome (CRS) > Gr 3 despite steroids. Blood samples were obtained at pre-dose and 0.5, 1, 3, 4.5, 6, 8, 24, and 48-hr after start of first bolus dose for pharmacokinetics (PK). Results: 26 pts with advanced solid tumors with a median age of 63 (44–75) yrs were enrolled. Median no. of doses was 7.5 (1–24). Table 1 outlines the PK parameters, DLTs and CRS. Due to a grade 5 CRS/death in cohort 3, the protocol was amended to include 20 mg IV dexamethasone prior to flavopiridol to prevent CRS (cohorts 2B, 1B). Of the 20 evaluable pts, 35% had stable- and 65% had progressive-disease. Results of serum cytokines (IL-2, IL-4, IL-6, TNF-a, IFN-g, IL-10) levels will be presented. Conclusions: There was a higher frequency of CRS, despite prophylactic steroids seen our pts with solid tumors compared to previous studies with CLL and this correlated with AUC. PK and toxicity profile in our pt population differs from pts with hematologic malignancies administered flavopiridol on the same schedule. Protein binding and serum albumin levels are under evaluation as potential contributors. This work is supported by NCI U01-CA76576. [Table: see text] [Table: see text]


Author(s):  
M. Archana ◽  
Mariya Palathingal ◽  
K. Athulya Damodharan ◽  
P. Ashisha ◽  
Nuaman, Akash Marathakam

Entrectinib is a selective inhibitor of tyrosine kinases , tropomyosin receptor kinases that targets oncogenic rearrangements in Neurotropic Tyrosine Receptor kinase, c-ros oncogene 1 and Anaplastic lymphoma kinase used for the treatment of various solid tumors. Entrectinib gained its first worldwide approval in Japan in June 2019 for the treatment of NTRK fusion-positive, advanced or recurring solid tumours in adults and children. In august 15, 2019 drug got FDA approval for the treatment of solid tumors in adult and children aged 12 and above. This article summarizes current status of Entrectinib from ongoing clinical trails and ideal place for drug in therapy.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 289-289
Author(s):  
Charles L. Bennett ◽  
Jessica A. Zagory ◽  
June M. McKoy ◽  
Kathryn R. McCaffrey ◽  
E. Allison Lyons ◽  
...  

Abstract Background: At ASH 2004 we reported that prior to 2004, eight drugs had received accelerated approval (AA) for ten hematologic oncology indications, only one had converted to full approval, and three serious adverse events associated with two of these drugs were identified post-approval. Following our presentation, Congressman Ed Markey (D-Massachussets) in June 2005 issued a report (“Conspiracy of Silence”) that raised similar concerns about the low rates of conversion from accelerated to regular approval and the failure of the FDA in ensuring that drug companies complete the required post-marketing studies. We update the experience with accelerated FDA approval for hematologic oncology. Methods: Information was obtained from the FDA on new drug applications and supplements for new uses, package inserts, and medical literature reviews. Results: Of 11 drug indications that received AA for hematologic oncology between 1995 and 2005, AA was generally granted for 9 based on clinical trials with &lt;100 patients that identified high response rates. In the past year, none have applied for conversion to regular approval. Bortezomib received regular approval for an indication different than that granted for AA. Three serious adverse drug reactions were identified for two indications (gemtuzumab ozogamicin, imatinib for GIST) within 2 years of AA approval. Conclusion: Since 2004, one new drug received AA for a hematologic oncology indication, one drug received regular approval for an indication different from that indicated under AA, and although 1 more year has elapsed, conversion has yet to occur for nine AA hematologic oncology indications. It is imperative that the FDA establish clear milestones regarding conversion to regular approval at the time AA is granted. Drug Name (AA Date; Years Lapsed Since AA) AA Indication Years to AA/Regular Approval N in Trial (Overall Response Rate) *Regular approval granted for myeloma failing one treatment Denileukin difitox (2/99; 6.5) T-cell lymphoma 1.17/- 71 (30%) Cytarabine liposomal (4/99; 6.3) Lymphomatous meningitis 0.46/- 14 (71%) Gemtuzumab ozogamicin (5/00; 5.35) CD33 positive AML 0.58/- 142 (30%) Alemtuzumab (5/01; 4.3) CLL 1.375/- 93 (33%) Imatinib mesylate (5/01) CML 0.25/2 864 (80%) Ibritumomab tuixetan (2/02; 3.5) Low-grade NHL 1.3/- 157(68%) Imatinib mesylate (2/02; 3.5) GIST 0.29/- 147 (54%) Imatinib mesylate (12/02; 2.7) CML peds 0.5/- 39 (67%) Bortezomib (5/03) Multiple myeloma w/ 2 prior chemo treatments* 0.46/2 193 (35%) Tositumomab (6/03/2.2) NHL 4.0/- 59 (71%) Clofarabine (12/04; 0.7) Relapsed/refractory ALL peds 0.75/- 49 (20%)


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3512-3512
Author(s):  
Renee C Prins ◽  
Stephen Spurgeon ◽  
Jeffrey W Tyner ◽  
Luke B Fletcher ◽  
Abdusebur Jemal ◽  
...  

Abstract Abstract 3512 Background: CK2 is a highly conserved and constitutively active serine-threonine protein kinase that plays a fundamental role in maintaining cell survival through pro-proliferative, anti-apoptotic and pro-angiogenic signaling. It is comprised of two catalytic (α and α') and two regulatory (β) subunits. Overexpression and hyperactivation of CK2 has been well described in a variety of human solid tumors, and more recently, in hematologic malignancies including chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML), T-cell acute lymphocytic leukemia (T-ALL), and multiple myeloma (MM). CK2 is known to support the viability of cancer cells through increased activation of the downstream PI3K/AKT signaling pathway, which results in cell-cycle progression, and suppression of apoptosis. Inhibition of CK2 has been shown to lead to preferential apoptosis of CLL, AML and myeloma cells. Studies have also demonstrated that CK2 inhibition in MM enhances the cytotoxic effect of chemotherapeutic drugs. Therefore, CK2 appears to be a rational target for novel therapies to treat hematologic malignancies. CX-4945 is a potent and orally bioavailable inhibitor of both isoforms of the CK2 catalytic subunits, and is the first small molecule inhibitor of CK2 to progress to human clinical trials. CX-4945 has been found to be highly selective for CK2 and has also been shown to inhibit phosphorylation of the CK2-specific S129 site of AKT and inhibit activation of the AKT pathway. Anti-tumor activity of CX-4945 has been validated in cancer cell lines and murine xenograft models, and biological activity was observed in early clinical trials with solid tumors and multiple myeloma. However, little is known about its possible effects in leukemias and lymphomas. Therefore, we explored the therapeutic potential of CX-4945 in 224 adult and pediatric patient samples across a wide spectrum of hematologic malignancies including AML, ALL, CLL, CML, CMML and leukemic phase lymphomas. Methods: Fresh primary cells from 224 patients were purified using a Ficoll gradient. Purified cells were then added to wells (5 × 104 per well) containing 10% serum containing media and four serial dilutions of CX-4945 (ranging from 10 nM to 10 μM). Three days after culturing cells in the presence or absence of each drug concentration, we performed a tetrazolium-based cell viability assay (MTS) to evaluate the effect of CX-4945 across a broad spectrum of primary hematologic malignancy samples. The viability data were normalized to untreated controls and used to calculate IC50 values. Results: Of the 7 categories of hematologic malignancies evaluated, all except for 2 (CMML and CML) demonstrated a subset of patients with significantly decreased viability (IC50 < 1 μM) in response to CX-4945 (Table 1). Investigations evaluating the effect of CK2 on downstream targets such as AKT and other cellular protein kinases, as well as studies exploring CK2 expression in AML are ongoing and will be presented. Conclusions: Our data confirms that CX-4945 demonstrates significant pre-clinical activity across a broad range of hematologic malignancies, and also confirms previous findings that CK2 may play a significant role in the tumorigenesis of certain hematologic malignancies. CX-4945 may be an attractive compound for clinical development, particularly in lymphoid malignancies such as CLL. Future studies will focus on the identification of highly sensitive subsets of cells, further defining the mechanistic details of the CK2 pathway in hematologic malignancies, and exploring the specific features that drive response to CK2 inhibition. Disclosures: Druker: Cylene Pharmaceuticals: Consultancy. Loriaux:Cylene Pharmaceuticals: Research Funding.


2020 ◽  
Vol 04 (04) ◽  
pp. 345-350
Author(s):  
Ryan J. Slovak ◽  
Hyun S. Kim

AbstractThe reinfusion of autologous or allogeneic immune cells that have been educated and/or engineered ex vivo to respond to tumor-specific antigens is termed “adoptive cell therapy.” While adoptive cell therapy has made tremendous strides in the treatment of hematologic malignancies, its utilization for solid tumors has lagged somewhat behind. The purpose of this article is to concisely review the clinical research that has been done to investigate adoptive cell therapy as a treatment for gastrointestinal malignancies.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Julien Edeline ◽  
Roch Houot ◽  
Aurélien Marabelle ◽  
Marion Alcantara

AbstractChimeric antigen receptor (CAR)-modified T cells and BiTEs are both immunotherapies which redirect T cell specificity against a tumor-specific antigen through the use of antibody fragments. They demonstrated remarkable efficacy in B cell hematologic malignancies, thus paving the way for their development in solid tumors. Nonetheless, the use of such new drugs to treat solid tumors is not straightforward. So far, the results from early phase clinical trials are not as impressive as expected but many improvements are under way. In this review we present an overview of the clinical development of CAR-T cells and BiTEs targeting the main antigens expressed by solid tumors. We emphasize the most frequent hurdles encountered by either CAR-T cells or BiTEs, or both, and summarize the strategies that have been proposed to overcome these obstacles.


Sign in / Sign up

Export Citation Format

Share Document