1
|
Garajová I, Peroni M, Gelsomino F, Leonardi F. A Simple Overview of Pancreatic Cancer Treatment for Clinical Oncologists. Curr Oncol 2023; 30:9587-9601. [PMID: 37999114 PMCID: PMC10669959 DOI: 10.3390/curroncol30110694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Pancreatic cancer (PDAC) is one of the most aggressive solid tumors and is showing increasing incidence. The aim of our review is to provide practical help for all clinical oncologists and to summarize the current management of PDAC using a simple "ABC method" (A-anatomical resectability, B-biological resectability and C-clinical conditions). For anatomically resectable PDAC without any high-risk factors (biological or conditional), the actual standard of care is represented by surgery followed by adjuvant chemotherapy. The remaining PDAC patients should all be treated with initial systemic therapy, though the intent for each is different: for borderline resectable patients, the intent is neoadjuvant; for locally advanced patients, the intent is conversion; and for metastatic PDAC patients, the intent remains just palliative. The actual standard of care in first-line therapy is represented by two regimens: FOLFIRINOX and gemcitabine/nab-paclitaxel. Recently, NALIRIFOX showed positive results over gemcitabine/nab-paclitaxel. There are limited data for maintenance therapy after first-line treatment, though 5-FU or FOLFIRI after initial FOLFIRINOX, and gemcitabine, after initial gemcitabine/nab-paclitaxel, might be considered. We also dedicate space to special rare conditions, such as PDAC with germline BRCA mutations, pancreatic acinar cell carcinoma and adenosquamous carcinoma of the pancreas, with few clinically relevant remarks.
Collapse
Affiliation(s)
- Ingrid Garajová
- Medical Oncology Unit, University Hospital of Parma, 43125 Parma, Italy; (M.P.)
| | - Marianna Peroni
- Medical Oncology Unit, University Hospital of Parma, 43125 Parma, Italy; (M.P.)
| | - Fabio Gelsomino
- Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Francesco Leonardi
- Medical Oncology Unit, University Hospital of Parma, 43125 Parma, Italy; (M.P.)
| |
Collapse
|
2
|
Strickler JH, Satake H, George TJ, Yaeger R, Hollebecque A, Garrido-Laguna I, Schuler M, Burns TF, Coveler AL, Falchook GS, Vincent M, Sunakawa Y, Dahan L, Bajor D, Rha SY, Lemech C, Juric D, Rehn M, Ngarmchamnanrith G, Jafarinasabian P, Tran Q, Hong DS. Sotorasib in KRAS p.G12C-Mutated Advanced Pancreatic Cancer. N Engl J Med 2023; 388:33-43. [PMID: 36546651 PMCID: PMC10506456 DOI: 10.1056/nejmoa2208470] [Citation(s) in RCA: 170] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND KRAS p.G12C mutation occurs in approximately 1 to 2% of pancreatic cancers. The safety and efficacy of sotorasib, a KRAS G12C inhibitor, in previously treated patients with KRAS p.G12C-mutated pancreatic cancer are unknown. METHODS We conducted a single-group, phase 1-2 trial to assess the safety and efficacy of sotorasib treatment in patients with KRAS p.G12C-mutated pancreatic cancer who had received at least one previous systemic therapy. The primary objective of phase 1 was to assess safety and to identify the recommended dose for phase 2. In phase 2, patients received sotorasib at a dose of 960 mg orally once daily. The primary end point for phase 2 was a centrally confirmed objective response (defined as a complete or partial response). Efficacy end points were assessed in the pooled population from both phases and included objective response, duration of response, time to objective response, disease control (defined as an objective response or stable disease), progression-free survival, and overall survival. Safety was also assessed. RESULTS The pooled population from phases 1 and 2 consisted of 38 patients, all of whom had metastatic disease at enrollment and had previously received chemotherapy. At baseline, patients had received a median of 2 lines (range, 1 to 8) of therapy previously. All 38 patients received sotorasib in the trial. A total of 8 patients had a centrally confirmed objective response (21%; 95% confidence interval [CI], 10 to 37). The median progression-free survival was 4.0 months (95% CI, 2.8 to 5.6), and the median overall survival was 6.9 months (95% CI, 5.0 to 9.1). Treatment-related adverse events of any grade were reported in 16 patients (42%); 6 patients (16%) had grade 3 adverse events. No treatment-related adverse events were fatal or led to treatment discontinuation. CONCLUSIONS Sotorasib showed anticancer activity and had an acceptable safety profile in patients with KRAS p.G12C-mutated advanced pancreatic cancer who had received previous treatment. (Funded by Amgen and others; CodeBreaK 100 ClinicalTrials.gov number, NCT03600883.).
Collapse
Affiliation(s)
- John H Strickler
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Hironaga Satake
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Thomas J George
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Rona Yaeger
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Antoine Hollebecque
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Ignacio Garrido-Laguna
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Martin Schuler
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Timothy F Burns
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Andrew L Coveler
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Gerald S Falchook
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Mark Vincent
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Yu Sunakawa
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Laetitia Dahan
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - David Bajor
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Sun-Young Rha
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Charlotte Lemech
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Dejan Juric
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Marko Rehn
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Gataree Ngarmchamnanrith
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Pegah Jafarinasabian
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - Qui Tran
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| | - David S Hong
- From Duke University Medical Center, Durham, NC (J.H.S.); Kansai Medical University, Shinmachi, Hirakata (H.S.), and St. Marianna University School of Medicine, Kawasaki (Y.S.) - both in Japan; University of Florida, Gainesville (T.J.G.); Memorial Sloan Kettering Cancer Center, New York (R.Y.); Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif (A.H.), and Marseille University Hospital, Marseille (L.D.) - both in France; Huntsman Cancer Institute, University of Utah, Salt Lake City (I.G.-L.); West German Cancer Center, University Hospital Essen, Essen (M.S.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (T.F.B.); Fred Hutchinson Cancer Center, University of Washington, Seattle (A.L.C.); Sarah Cannon Research Institute at HealthONE, Denver (G.S.F.); London Regional Cancer Program, London, ON, Canada (M.V.); University Hospitals Cleveland Medical Center, Cleveland (D.B.); Yonsei Cancer Center, Seoul, South Korea (S.-Y.R.); Scientia Clinical Research and Prince of Wales Clinical School, University of New South Wales, Sydney (C.L.); Massachusetts General Cancer Center, Boston (D.J.); Amgen, Thousand Oaks, CA (M.R., G.N., P.J., Q.T.); and University of Texas M.D. Anderson Cancer Center, Houston (D.S.H.)
| |
Collapse
|
3
|
Noel MS, Kim S, Hartley ML, Wong S, Picozzi V, Staszewski H, Kim DW, Van Tornout JM, Philip PA, Chung V, Ocean AJ, Wang‐Gillam A. A randomized phase II study of SM-88 plus methoxsalen, phenytoin, and sirolimus in patients with metastatic pancreatic cancer treated in the second line and beyond. Cancer Med 2022; 11:4169-4181. [PMID: 35499204 PMCID: PMC9678093 DOI: 10.1002/cam4.4768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/11/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This trial explores SM-88 used with methoxsalen, phenytoin, and sirolimus (MPS) in pretreated metastatic pancreatic ductal adenocarcinoma (mPDAC) METHODS: Forty-nine patients were randomized to daily 460 or 920 mg oral SM-88 with MPS (SM-88 Regimen). The primary endpoint was objective response rate (RECIST 1.1). RESULTS Thirty-seven patients completed ≥ one cycle of SM-88 Regimen (response evaluable population). Disease control rate (DCR), overall survival (OS), and progression-free survival (PFS) did not differ significantly between dose levels. Stable disease was achieved in 9/37 patients (DCR, 24.3%); there were no complete or partial responses. Quality-of-life (QOL) was maintained and trended in favor of 920 mg. SM-88 Regimen was well tolerated; a single patient (1/49) had related grade 3 and 4 adverse events, which later resolved. In the intention-to-treat population of 49 patients, the median overall survival (mOS) was 3.4 months (95% CI: 2.7-4.9 months). Those treated in the second line had an mOS of 8.1 months and a median PFS of 3.8 months. Survival was higher for patients with stable versus progressive disease (any line; mOS: 10.6 months vs. 3.9 months; p = 0.01). CONCLUSIONS SM-88 Regimen has a favorable safety profile with encouraging QOL effects, disease control, and survival trends. This regimen should be explored in the second-line treatment of patients with mPDAC. CLINICALTRIALS gov Identifier: NCT03512756.
Collapse
Affiliation(s)
- Marcus S. Noel
- Georgetown Lombardi Comprehensive Cancer CenterWashingtonDistrict of ColumbiaUSA
| | - Semmie Kim
- TYME Technologies Inc.BedminsterNew JerseyUSA
| | - Marion L. Hartley
- The Ruesch Center for the Cure of Gastrointestinal CancersWashingtonDistrict of ColumbiaUSA
| | - Steve Wong
- Sarcoma Oncology Research CenterSanta MonicaCaliforniaUSA
| | | | | | - Dae Won Kim
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Philip Agop Philip
- Karmanos Cancer CenterWayne State UniversityMichiganDetroitUSA
- SWOGFarmington HillsMichiganUSA
| | | | - Allyson J. Ocean
- Weill Cornell MedicineNew York‐Presbyterian HospitalNew YorkNew YorkUSA
| | - Andrea Wang‐Gillam
- Washington University School of Medicine in St. LouisSt. LouisMissouriUSA
| |
Collapse
|
4
|
Chakrabarti S, Kamgar M, Mahipal A. Systemic Therapy of Metastatic Pancreatic Adenocarcinoma: Current Status, Challenges, and Opportunities. Cancers (Basel) 2022; 14:2588. [PMID: 35681565 PMCID: PMC9179239 DOI: 10.3390/cancers14112588] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by nonspecific presenting symptoms, lack of a screening test, rapidly progressive clinical course, and presentation with an advanced-stage disease in the majority of patients. PDAC is essentially a systemic disease irrespective of the initial stage, as most patients with non-metastatic PDAC undergoing curative-intent treatment eventually experience metastatic relapse. Currently, cytotoxic chemotherapy remains the cornerstone of treatment in patients with advanced disease. However, the current standard treatment with multiagent chemotherapy has modest efficacy and results in median overall survival (OS) of less than a year and a 5-year OS of about 10%. The pathobiology of PDAC poses many challenges, including a unique tumor microenvironment interfering with drug delivery, intratumoral heterogeneity, and a strongly immunosuppressive microenvironment that supports cancer growth. Recent research is exploring a wide range of novel therapeutic targets, including genomic alterations, tumor microenvironment, and tumor metabolism. The rapid evolution of tumor genome sequencing technologies paves the way for personalized, targeted therapies. The present review summarizes the current chemotherapeutic treatment paradigm of advanced PDAC and discusses the evolving novel targets that are being investigated in a myriad of clinical trials.
Collapse
Affiliation(s)
- Sakti Chakrabarti
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (S.C.); (M.K.)
| | - Mandana Kamgar
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (S.C.); (M.K.)
| | - Amit Mahipal
- Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
5
|
An In Vitro Investigation into Cryoablation and Adjunctive Cryoablation/Chemotherapy Combination Therapy for the Treatment of Pancreatic Cancer Using the PANC-1 Cell Line. Biomedicines 2022; 10:biomedicines10020450. [PMID: 35203660 PMCID: PMC8962332 DOI: 10.3390/biomedicines10020450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
As the incidence of pancreatic ductal adenocarcinoma (PDAC) continues to grow, so does the need for new strategies for treatment. One such area being evaluated is cryoablation. While promising, studies remain limited and questions surrounding basic dosing (minimal lethal temperature) coupled with technological issues associated with accessing PDAC tumors and tumor proximity to vasculature and bile ducts, among others, have limited the use of cryoablation. Additionally, as chemotherapy remains the first-line of attack for PDAC, there is limited information on the impact of combining freezing with chemotherapy. As such, this study investigated the in vitro response of a PDAC cell line to freezing, chemotherapy, and the combination of chemotherapy pre-treatment and freezing. PANC-1 cells and PANC-1 tumor models were exposed to cryoablation (freezing insult) and compared to non-frozen controls. Additionally, PANC-1 cells were exposed to varying sub-clinical doses of gemcitabine or oxaliplatin alone and in combination with freezing. The results show that freezing to −10 °C did not affect viability, whereas −15 °C and −20 °C resulted in a reduction in 1 day post-freeze viability to 85% and 20%, respectively, though both recovered to controls by day 7. A complete cell loss was found following a single freeze below −25 °C. The combination of 100 nM gemcitabine (1.1 mg/m2) pre-treatment and a single freeze at −15 °C resulted in near-complete cell death (<5% survival) over the 7-day assessment interval. The combination of 8.8 µM oxaliplatin (130 mg/m2) pre-treatment and a single −15 °C freeze resulted in a similar trend of increased PANC-1 cell death. In summary, these in vitro results suggest that freezing alone to temperatures in the range of −25 °C results in a high degree of PDAC destruction. Further, the data support a potential combinatorial chemo/cryo-therapeutic strategy for the treatment of PDAC. These results suggest that a reduction in chemotherapeutic dose may be possible when offered in combination with freezing for the treatment of PDAC.
Collapse
|
6
|
Barros AG, Pulido CF, Machado M, Brito MJ, Couto N, Sousa O, Melo SA, Mansinho H. Treatment optimization of locally advanced and metastatic pancreatic cancer (Review). Int J Oncol 2021; 59:110. [PMID: 34859257 PMCID: PMC8651228 DOI: 10.3892/ijo.2021.5290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant tumor types, being the sixth leading cause of mortality worldwide and the fourth in Europe. Globally, it has a mortality/incidence ratio of 98%, and the 5‑year survival rate in Europe is only 3%. Although risk factors, such as obesity, diabetes mellitus, smoking, alcohol consumption and genetic factors, have been identified, the causes of PDAC remain elusive. Additionally, the only curative treatment for PDAC is surgery with negative margins. However, upon diagnosis, ~30% of the patients already present with locally advanced disease. In these cases, a multidisciplinary approach is required to improve disease‑related symptoms and prolong patient survival. In the present article, a comprehensive review of PDAC epidemiology, physiology and treatment is provided. Moreover, guidelines on patient treatment are suggested. Among the different available therapeutic options for the treatment of advanced PDAC, results are modest, most likely due to the complexity of the disease, and so the prognostic remains poor. Molecular approaches based on multi‑omics research are promising and will contribute to groundbreaking personalized medicine. Thus, economic investment that promotes research of pancreatic cancer will be critical to the development of more efficient diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Anabela G. Barros
- Department of Medical Oncology, University Hospital of Coimbra, 3004-561 Coimbra, Portugal
| | - Catarina F. Pulido
- Department of Medical Oncology, Luz Lisbon Hospital, 1500-650 Lisbon, Portugal
| | - Manuela Machado
- Department of Medical Oncology, Entre o Douro e Vouga Hospital Center (CHEDV), 4520-211 Santa Maria da Feira, Portugal
| | - Maria José Brito
- Pathologic Anatomy Department, Garcia de Orta Hospital, 2805-267 Almada, Portugal
| | - Nuno Couto
- Digestive Unit, Champalimaud Clinical Centre, 4200-135 Porto, Portugal
- Champalimaud Research Centre, 1400-038 Lisbon, 4200-135 Porto, Portugal
| | - Olga Sousa
- Radiotherapy Department, Portuguese Institute of Oncology, 4200-072 Porto, 4200-135 Porto, Portugal
| | - Sónia A. Melo
- i3S-Institute for Research and Innovation in Health of University of Porto, 4200-135 Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hélder Mansinho
- Hemato-Oncology Department, Garcia de Orta Hospital, 2805-267 Almada, Portugal
| |
Collapse
|
7
|
Kamgar M, Chakrabarti S, Shreenivas A, George B. Evolution of Systemic Therapy in Metastatic Pancreatic Ductal Adenocarcinoma. Surg Oncol Clin N Am 2021; 30:673-691. [PMID: 34511189 DOI: 10.1016/j.soc.2021.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pancreatic ductal adenocarcinoma is characterized by early systemic dissemination, a complex tumor microenvironment, as well as significant intratumoral and intertumoral heterogeneity. Treatment options and survival in pancreatic ductal adenocarcinoma have improved steadily over the last 3 decades. Although cytotoxic chemotherapy is currently the mainstay of treatment for pancreatic ductal adenocarcinoma, evolving therapeutic strategies are aimed at targeting the tumor microenvironment, metabolism, and the tumor-host immune balance.
Collapse
Affiliation(s)
- Mandana Kamgar
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA.
| | - Sakti Chakrabarti
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| | - Aditya Shreenivas
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| | - Ben George
- Division of Hematology and Oncology, Department of Medicine, LaBahn Pancreatic Cancer Program, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA
| |
Collapse
|
8
|
Hani U, Osmani RAM, Siddiqua A, Wahab S, Batool S, Ather H, Sheraba N, Alqahtani A. A systematic study of novel drug delivery mechanisms and treatment strategies for pancreatic cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Gutierrez-Sainz L, Viñal D, Villamayor J, Martinez-Perez D, Garcia-Cuesta JA, Ghanem I, Custodio A, Feliu J. Prognostic factors in advanced pancreatic ductal adenocarcinoma patients-receiving second-line treatment: a single institution experience. Clin Transl Oncol 2021; 23:1838-1846. [PMID: 33866520 DOI: 10.1007/s12094-021-02589-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Second-line (2L) treatments for advanced pancreatic ductal adenocarcinoma (PDAC) achieve a modest benefit at the expense of potential toxicity. In the absence of predictive factors of response, the identification of prognostic factors could help in the therapeutic decisions-making. The purpose of this study was to assess the prognostic factors associated with shorter survival in patients with advanced PDAC who received 2L treatment. METHODS We conducted a single institution retrospective study, which included all patients with advanced PDAC who received 2L treatment between September 2006 and February 2020 at La Paz University Hospital, Madrid (Spain). Significant variables in the logistic regression model were used to create a prognostic score. RESULTS We included 108 patients. The median overall survival (OS) was 5.10 months (95%CI 4.02-6.17). In the multivariate analysis, time to progression (TTP) shorter than 4 months after first-line treatment (OR 4.53 [95%CI 1.28-16.00] p = 0.01), neutrophil-to-lymphocyte ratio (NLR) greater than 3 at the beginning of 2L (OR 9.07 [95%CI 1.82-45.16] p = 0.01) and CA-19.9 level higher than the upper limit of normal at the beginning of 2L (OR 7.83 [95%CI 1.30-49.97] p = 0.02) were independently associated with OS shorter than 3 months. The prognostic score classified patients into three prognostic groups (good, intermediate and poor) with significant differences in OS (p < 0.001). CONCLUSIONS TTP shorter than 4 months after first-line treatment, NLR greater than 3 and CA-19.9 level higher than the upper limit of normal at the beginning of 2L were associated with shorter overall survival. We developed a prognostic score that classifies patients with advanced PDAC into three prognostic groups after progression to the first-line. This score could help in the decision-making for 2L treatment.
Collapse
Affiliation(s)
- L Gutierrez-Sainz
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | - D Viñal
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - J Villamayor
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - D Martinez-Perez
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - J A Garcia-Cuesta
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - I Ghanem
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - A Custodio
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Cátedra UAM-AMGEN, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - J Feliu
- Medical Oncology Department, Hospital Universitario La Paz, IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Cátedra UAM-AMGEN, Madrid, Spain.,CIBERONC, Madrid, Spain
| |
Collapse
|
10
|
Okano N, Morizane C, Nomura S, Takahashi H, Tsumura H, Satake H, Mizuno N, Tsuji K, Shioji K, Asagi A, Yasui K, Kitagawa S, Kashiwada T, Ishiguro A, Kanai M, Ueno M, Ogura T, Shimizu S, Tobimatsu K, Motoya M, Nakashima K, Ikeda M, Okusaka T, Furuse J. Phase II clinical trial of gemcitabine plus oxaliplatin in patients with metastatic pancreatic adenocarcinoma with a family history of pancreatic/breast/ovarian/prostate cancer or personal history of breast/ovarian/prostate cancer (FABRIC study). Int J Clin Oncol 2020; 25:1835-1843. [PMID: 32535711 DOI: 10.1007/s10147-020-01721-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/02/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND A family/personal history of breast, ovarian, or pancreatic cancer is a useful predictive marker for response to platinum-based chemotherapy in treating patients with pancreatic cancer. These cancers, and prostate cancer, are known as BRCA-related malignancies. We evaluated the efficacy of gemcitabine plus oxaliplatin (GEMOX) in patients with metastatic pancreatic cancer with a family/personal history of these cancers. METHODS Chemotherapy-naïve patients with metastatic pancreatic cancer with a family history of pancreatic/breast/ovarian/prostate cancer or a personal history of breast/ovarian/prostate cancer were included. Patients received fixed dose-rate gemcitabine (1000 mg/m2) and oxaliplatin (100 mg/m2) every 2 weeks. The primary endpoint was 1-year survival, and the threshold and expected values were set at 30 and 50%, respectively. The target sample size was determined to be 43, with a one-sided alpha value of 5% and power of 80%. A total of 45 patients were enrolled. RESULTS Among the first 43 enrolled patients, the 1-year survival rate was 27.9% [90% confidence interval (CI) 17.0-41.3], which did not meet the primary endpoint. Median overall survival, progression-free survival, and response rates were 7.6 months (95% CI 6.0-10.7), 4.0 months (95% CI 2.0-4.6), and 26.7% (95% CI 14.6-41.9), respectively, in all registered patients. The GEMOX regimen was generally tolerated; the most common grade three or higher adverse events were hematological toxicities. CONCLUSION GEMOX did not show the expected efficacy in patients with metastatic pancreatic cancer with a family or personal history of pancreatic/breast/ovarian/prostate cancer. Selection of GEMOX based on family/personal history is not recommended. TRIAL REGISTRATION NUMBER UMIN000017894.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan.
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shogo Nomura
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Kashiwa, Japan
| | - Hideaki Takahashi
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hidetaka Tsumura
- Department of Gastroenterology, Hyogo Cancer Center, Hyogo, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Kunihiro Tsuji
- Department of Gastroenterology, Ishikawa Prefectural Central Hospital, Ishikawa, Japan
| | - Kazuhiko Shioji
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | - Akinori Asagi
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Kohichiroh Yasui
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sho Kitagawa
- Department of Gastroenterology, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Tomomi Kashiwada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Atsushi Ishiguro
- Department of Medical Oncology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Masashi Kanai
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, Japan
| | - Takashi Ogura
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Kazutoshi Tobimatsu
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masayo Motoya
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koji Nakashima
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
De Jesus-Acosta A, Narang A, Mauro L, Herman J, Jaffee EM, Laheru DA. Carcinoma of the Pancreas. ABELOFF'S CLINICAL ONCOLOGY 2020:1342-1360.e7. [DOI: 10.1016/b978-0-323-47674-4.00078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Tesfaye AA, Wang H, Hartley ML, He AR, Weiner L, Gabelia N, Kapanadze L, Shezad M, Brody JR, Marshall JL, Pishvaian MJ. A Pilot Trial of Molecularly Tailored Therapy for Patients with Metastatic Pancreatic Ductal Adenocarcinoma. J Pancreat Cancer 2019; 5:12-21. [PMID: 31065624 PMCID: PMC6503449 DOI: 10.1089/pancan.2019.0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose: Despite the wide adoption of tumor molecular profiling, there is a dearth of evidence linking molecular biomarkers for treatment selection to prediction of treatment outcomes in patients with metastatic pancreatic cancer. We initiated a pilot study to test the feasibility of designing a larger phase II trial of molecularly tailored treatment for metastatic pancreatic cancer. Methods: Our study aimed to assess the feasibility of following a treatment algorithm based on the expression of three published predictive markers of response to chemotherapy: ribonucleotide reductase catalytic subunit M1 (for gemcitabine); excision repair cross-complementation group 1 (for platinum agents); and thymidylate synthase (for 5-fluorouracil) in patients with untreated, metastatic pancreatic cancer. Results of the tumor biopsy analysis were used to assign patients to one of seven doublet regimens. Key secondary objectives included response rate (RR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Results: Between December 2012 and March 2015, 30 patients were enrolled into the study. Ten patients failed screening primarily due to inadequate tumor tissue availability. Of the remaining 20 patients, 19 were assigned into 6 different chemotherapy doublets, and achieved an RR of 28%, with a DCR rate of 78%. The median PFS and OS were 5.78 and 8.21 months, respectively. Conclusions: The incorporation of biomarkers into a treatment algorithm is feasible and resulted in a PFS and OS similar to other doublet therapies for patients with metastatic pancreatic cancer. Based on the results from this pilot study, a larger phase II randomized trial of molecularly targeted therapy versus physicians' choice of standard of care has been initiated in the second-line setting (NCT02967770).
Collapse
Affiliation(s)
- Anteneh A Tesfaye
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Hongkun Wang
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Marion L Hartley
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Aiwu Ruth He
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Louis Weiner
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Nina Gabelia
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Lana Kapanadze
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Muhammad Shezad
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Jonathan R Brody
- Department of Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - John L Marshall
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Michael J Pishvaian
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| |
Collapse
|
13
|
El-Zahaby SA, Elnaggar YSR, Abdallah OY. Reviewing two decades of nanomedicine implementations in targeted treatment and diagnosis of pancreatic cancer: An emphasis on state of art. J Control Release 2019; 293:21-35. [PMID: 30445002 DOI: 10.1016/j.jconrel.2018.11.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/10/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is nowadays the most life-threatening cancer type worldwide. The problem of poor diagnosis, anti-neoplastics resistance and biopharmaceutical drawbacks of effective anti-cancer drugs lead to worsen disease state. Nanotechnology-based carrier systems used in both imaging and treatment procedures had solved many of these problems. It is critical to develop advanced detection method to save patients from being too late diagnosed. Targeting the pancreatic cancer cells as well helped in decreasing the side effects associated with normal cells destruction. Drug resistance is another challenge in pancreatic cancer management that can be solved by thorough understanding of the microenvironment associated with the disease to design creative nanocarriers. This is the first article to review multifaceted approaches of nanomedicine in pancreatic cancer detection and management. Additionally, mortality rates in selected Arab and European countries were illustrated herein. An emphasis was given on therapeutic and diagnostic challenges and different nanotechnologies adopted to overcome. The four main approaches encompassed nanomedicine for herbal treatment, nanomedicine of synthetic anti-cancer drugs, metal nanoparticles as a distinct treatment policy and nanotechnology for cancer diagnosis. Future research perspectives have been finally proposed.
Collapse
Affiliation(s)
- Sally A El-Zahaby
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
14
|
El-Zahaby SA, Elnaggar YS, Abdallah OY. Reviewing two decades of nanomedicine implementations in targeted treatment and diagnosis of pancreatic cancer: An emphasis on state of art. J Control Release 2019. [DOI: https://doi.org/10.1016/j.jconrel.2018.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
15
|
Olson JL, Bold RJ. Currently available first-line drug therapies for treating pancreatic cancer. Expert Opin Pharmacother 2018; 19:1927-1940. [PMID: 30325679 DOI: 10.1080/14656566.2018.1509954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic adenocarcinoma is the 9th most common cancer in the United States and the 4th most common cause of cancer-related death given its poor prognosis. AREAS COVERED The authors have performed a literature search for pertinent published clinical trials, ongoing Phase 3 clinical trials, and current treatment guidelines using PubMed, Clinicaltrials.gov, and NCCN, ASCO, ESMO, and JPS websites. The review itself discusses landmark studies and ongoing research into the chemotherapy regimens recommended by each oncologic society. The authors also examine drugs that were promising but failed in Phase 3 trials and those currently being investigated. Finally, the authors provide their expert opinion on the subject and provide their future perspectives. EXPERT OPINION While advances in chemotherapy for pancreatic cancer have been limited in comparison to other cancers, there have been improvements in survival. Combination therapy and a goal of R0 resection are key elements to extend life. Novel agents directed at the unique properties of pancreatic cancer are promising.
Collapse
Affiliation(s)
- Jennifer L Olson
- a Division of Surgical Oncology , UC Davis Cancer Center , Sacramento , CA , USA
| | - Richard J Bold
- a Division of Surgical Oncology , UC Davis Cancer Center , Sacramento , CA , USA
| |
Collapse
|
16
|
Hua J, Shi S, Liang D, Liang C, Meng Q, Zhang B, Ni Q, Xu J, Yu X. Current status and dilemma of second-line treatment in advanced pancreatic cancer: is there a silver lining? Onco Targets Ther 2018; 11:4591-4608. [PMID: 30122951 PMCID: PMC6084072 DOI: 10.2147/ott.s166405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer remains one of the most lethal malignant diseases worldwide. The majority of patients present with advanced disease and, therefore, need palliative chemotherapy. Some chemotherapeutic regimens have been well established as first-line therapies and have been shown to increase survival; however, almost all patients with advanced pancreatic cancer will experience disease progression after first-line therapy. Nevertheless, many patients who retain good performance status after initial treatment remain good candidates for additional therapy. Historically, few studies have assessed second-line therapy, with most reports representing small phase II trials with variable findings; however, clinical research for second-line treatment has increased in the past decade, and several randomized controlled trials using different regimens have been published. The current literature shows varying results on treatment efficacy and tolerability. Thus, we reviewed the published data on the use of chemotherapy in the second-line setting for the treatment of advanced pancreatic cancer.
Collapse
Affiliation(s)
- Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Dingkong Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, ; .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China, ; .,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China, ; .,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China, ;
| |
Collapse
|
17
|
Saletti P, Zaniboni A. Second-line therapy in advanced upper gastrointestinal cancers: current status and new prospects. J Gastrointest Oncol 2018; 9:377-389. [PMID: 29755778 DOI: 10.21037/jgo.2018.01.12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The prognosis of patients with advanced upper gastrointestinal cancers (UGC) remains poor. Current available systemic armamentarium is limited, and little progress has been made over the last decades. Main achievements have been obtained in first-line setting, however an increasingly proportion of patients are considered for second-line therapy, although data from randomized trials are scarce or even lacking. In this comprehensive review we examine the literature to summarize the efficacy and limitations of second-line systemic options in patients with advanced UGC, with a glimpse into the innovations.
Collapse
Affiliation(s)
- Piercarlo Saletti
- Medical Oncology Department, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Alberto Zaniboni
- Dipartimento Oncologico, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| |
Collapse
|
18
|
Uccello M, Moschetta M, Mak G, Alam T, Henriquez CM, Arkenau HT. Towards an optimal treatment algorithm for metastatic pancreatic ductal adenocarcinoma (PDA). ACTA ACUST UNITED AC 2018; 25:e90-e94. [PMID: 29507500 DOI: 10.3747/co.25.3708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotherapy remains the mainstay of treatment for advanced pancreatic ductal adenocarcinoma (pda). Two randomized trials have demonstrated superiority of the combination regimens folfirinox (5-fluorouracil, leucovorin, oxaliplatin, and irinotecan) and gemcitabine plus nab-paclitaxel over gemcitabine monotherapy as a first-line treatment in adequately fit subjects. Selected pda patients progressing to first-line therapy can receive secondline treatment with moderate clinical benefit. Nevertheless, the optimal algorithm and the role of combination therapy in second-line are still unclear. Published second-line pda clinical trials enrolled patients progressing to gemcitabine-based therapies in use before the approval of nab-paclitaxel and folfirinox. The evolving scenario in second-line may affect the choice of the first-line treatment. For example, nanoliposomal irinotecan plus 5-fluouracil and leucovorin is a novel second-line option which will be suitable only for patients progressing to gemcitabine-based therapy. Therefore, clinical judgement and appropriate patient selection remain key elements in treatment decision. In this review, we aim to illustrate currently available options and define a possible algorithm to guide treatment choice. Future clinical trials taking into account sequential treatment as a new paradigm in pda will help define a standard algorithm.
Collapse
Affiliation(s)
- M Uccello
- Drug Development, Sarah Cannon Research Institute UK, London, United Kingdom; and
| | - M Moschetta
- Drug Development, Sarah Cannon Research Institute UK, London, United Kingdom; and
| | - G Mak
- Drug Development, Sarah Cannon Research Institute UK, London, United Kingdom; and
| | - T Alam
- Drug Development, Sarah Cannon Research Institute UK, London, United Kingdom; and
| | - C Murias Henriquez
- Drug Development, Sarah Cannon Research Institute UK, London, United Kingdom; and
| | - H-T Arkenau
- Drug Development, Sarah Cannon Research Institute UK, London, United Kingdom; and.,UCL Cancer Institute, University College of London, London, United Kingdom
| |
Collapse
|
19
|
Adjuvant or Neoadjuvant Therapy in the Treatment in Pancreatic Malignancies: Where Are We? Surg Clin North Am 2017; 98:95-111. [PMID: 29191281 DOI: 10.1016/j.suc.2017.09.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Since the advent of modern surgery for pancreatic cancer, clinicians have recognized this cancer's propensity to recur locally, metastasize, and cause death. Despite significant efforts to improve patient outcomes with better adjuvant therapy, only modest gains in survival have been observed. An alternative strategy of neoadjuvant therapy followed by surgery has the potential to improve patient selection and survival, and expand the pool of patients eligible for curative surgery. This article summarizes large, randomized trials of adjuvant therapy, explains the limitations imposed by up-front surgery, and suggests neoadjuvant therapy as a rational alternative to initial surgery and adjuvant therapy.
Collapse
|
20
|
Bullock A, Stuart K, Jacobus S, Abrams T, Wadlow R, Goldstein M, Miksad R. Capecitabine and oxaliplatin as first and second line treatment for locally advanced and metastatic pancreatic ductal adenocarcinoma. J Gastrointest Oncol 2017; 8:945-952. [PMID: 29299353 DOI: 10.21037/jgo.2017.06.06] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background There are limited treatment options available for patients with advanced pancreatic ductal adenocarcinoma (PDAC). We conducted a phase II study evaluating the efficacy and safety of capecitabine/oxaliplatin (CAPOX) in patients with locally advanced and metastatic PDAC treated in the first and second lines. Methods Forty subjects with advanced PDAC and ECOG performance status ≥2 were enrolled. Treatment consisted of capecitabine 2,000 mg/m2 orally in two divided doses daily for 14 days and oxaliplatin 130 mg/m2 intravenously day 1 every 21 days. The primary endpoint was response rate (RR); secondary endpoints included safety analysis, progression free survival (PFS) and overall survival (OS). Results The overall RR was 12.5% (N=3); the disease control rate was 67% (N=16). Due to the protocol definition for eligibility of response evaluation, only 60% (N=24) were evaluable for the primary endpoint. Median progression free survival (mPFS) was 3.8 months (95% CI: 1.3, 6.2); median OS (mOS) was 7.4 months (95% CI: 4.8, 12.2). The most common grade 3/4 toxicities included: fatigue (19%), nausea (17%), and diarrhea (14%). Conclusions CAPOX is an active regimen in patients with advanced PDAC and is associated with acceptable toxicity. Careful consideration should be given to response endpoints and outcome measures when studying this characteristically ill population.
Collapse
Affiliation(s)
| | - Keith Stuart
- Lahey Hospital and Medical Center, Burlington, MA, USA
| | | | | | | | | | | |
Collapse
|
21
|
Chung KH, Ryu JK, Son JH, Lee JW, Jang DK, Lee SH, Kim YT. Efficacy of Capecitabine Plus Oxaliplatin Combination Chemotherapy for Advanced Pancreatic Cancer after Failure of First-Line Gemcitabine-Based Therapy. Gut Liver 2017; 11:298-305. [PMID: 27965478 PMCID: PMC5347656 DOI: 10.5009/gnl16307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Background/Aims Second-line chemotherapy in patients with advanced pancreatic ductal adenocarcinoma (PDAC) that progresses following gemcitabine-based treatment has not been established. This study aimed to investigate the efficacy and safety of second-line combination chemotherapy with capecitabine and oxaliplatin (XELOX) in these patients. Methods Between August 2011 and May 2014, all patients who received at least one cycle of XELOX (capecitabine, 1,000 mg/m2 twice daily for 14 days; oxaliplatin, 130 mg/m2 on day 1 of a 3-week cycle) combination chemotherapy for unresectable or recurrent PDAC were retrospectively recruited. The response was evaluated every 9 weeks, and the tumor response rate, progression-free survival and overall survival, and adverse events were assessed. Results Sixty-two patients were included; seven patients (11.3%) had a partial tumor response, and 20 patients (32.3%) had stable disease. The median progression-free and overall survival were 88 days (range, 35.1 to 140.9 days) and 158 days (range, 118.1 to 197.9 days), respectively. Patients who remained stable longer with frontline therapy (≥120 days) exhibited significantly longer progression-free and overall survival. The most common grade 3 to 4 adverse events in patients were vomiting (8.1%) and anorexia (6.5%). There was one treatment-related mortality caused by severe neutropenia and typhlitis. Conclusions Second-line XELOX combination chemotherapy demonstrated an acceptable response and survival rate in patients with advanced PDAC who had failed gemcitabine-based chemotherapy.
Collapse
Affiliation(s)
- Kwang Hyun Chung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Kon Ryu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jun Hyuk Son
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Woo Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Kee Jang
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong-Tae Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Ruess DA, Görgülü K, Wörmann SM, Algül H. Pharmacotherapeutic Management of Pancreatic Ductal Adenocarcinoma: Current and Emerging Concepts. Drugs Aging 2017; 34:331-357. [PMID: 28349415 DOI: 10.1007/s40266-017-0453-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma is a devastating malignancy, which is the result of late diagnosis, aggressive disease, and a lack of effective treatment options. Thus, pancreatic ductal adenocarcinoma is projected to become the second leading cause of cancer-related death by 2030. This review summarizes recent developments of oncological therapy in the palliative setting of metastatic pancreatic ductal adenocarcinoma. It further compiles novel targets and therapeutic approaches as well as promising treatment combinations, which are presently in preclinical evaluation, covering several aspects of the hallmarks of cancer. Finally, challenges to the implementation of an individualized therapy approach in the context of precision medicine are discussed.
Collapse
Affiliation(s)
- Dietrich A Ruess
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Kivanc Görgülü
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Sonja M Wörmann
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Hana Algül
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
23
|
Perales-Patón J, Piñeiro-Yañez E, Tejero H, López-Casas PP, Hidalgo M, Gómez-López G, Al-Shahrour F. Pancreas Cancer Precision Treatment Using Avatar Mice from a Bioinformatics Perspective. Public Health Genomics 2017; 20:81-91. [PMID: 28858862 DOI: 10.1159/000479812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 01/03/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related death among solid malignancies. Unfortunately, PDAC lethality has not substantially decreased over the past 20 years. This aggressiveness is related to the genomic complexity and heterogeneity of PDAC, but also to the absence of an effective screening for the detection of early-stage tumors and a lack of efficient therapeutic options. Therefore, there is an urgent need to improve the arsenal of anti-PDAC drugs for an effective treatment of these patients. Patient-derived xenograft (PDX) mouse models represent a promising strategy to personalize PDAC treatment, offering a bench testing of candidate treatments and helping to select empirical treatments in PDAC patients with no therapeutic targets. Moreover, bioinformatics-based approaches have the potential to offer systematic insights into PDAC etiology predicting putatively actionable tumor-specific genomic alterations, identifying novel biomarkers and generating disease-associated gene expression signatures. This review focuses on recent efforts to individualize PDAC treatments using PDX models. Additionally, we discuss the current understanding of the PDAC genomic landscape and the putative druggable targets derived from mutational studies. PDAC molecular subclassifications and gene expression profiling studies are reviewed as well. Finally, latest bioinformatics methodologies based on somatic variant detection and prioritization, in silico drug response prediction, and drug repositioning to improve the treatment of advanced PDAC tumors are also covered.
Collapse
Affiliation(s)
- Javier Perales-Patón
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
24
|
Petrelli F, Inno A, Ghidini A, Rimassa L, Tomasello G, Labianca R, Barni S. Second line with oxaliplatin- or irinotecan-based chemotherapy for gemcitabine-pretreated pancreatic cancer: A systematic review. Eur J Cancer 2017. [PMID: 28633088 DOI: 10.1016/j.ejca.2017.05.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
BACKGROUND oxaliplatin (OXA)- and irinotecan (IRI)-based chemotherapies are the most frequently used salvage regimens in patients with metastatic pancreatic cancer (PC) after first-line gemcitabine-based therapy. There are no prospective comparisons of these regimens in this setting. We conducted a systematic review of published trials to compare the efficacy of these treatments. METHODS studies that enrolled patients with stage IV disease receiving chemotherapy with OXA or IRI plus fluoropyrimidines were identified using electronic databases (Pubmed, Embase, SCOPUS, CINAHL, Web of Science and Cochrane Library). Clinical outcomes were compared using weighted values of median overall survival (OS), progression-free survival (PFS), response rates (RRs), and clinical benefit rates (CBRs). A 2-tailed t-test with a significance level of 0.05 for comparisons of continuous variables and a Chi-squared test for comparisons of proportions were used. RESULTS overall, 24 studies were included. The pooled overall response rate (ORR), disease control rate (DCR), PFS and OS were 11%, 37.9%, 2.87 and 5.48 months respectively. There was no significant difference in response rates between OXA-based and IRI-based chemotherapies (11.9% versus 8.7%; Chi-squared P = 0.1), respectively. Also there was no significant difference in median PFS (2.9 months versus 2.7 months; t-test P = 0.72), OS (5.3 months versus 5.5 months; t-test P = 0.72), but a greater DCR with OXA-based chemotherapy (41.1% versus 29.4%; Chi-squared P = 0.0008). CONCLUSION OXA- and IRI-containing regimens were associated with similar efficacy when used after gemcitabine-based chemotherapy in patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Fausto Petrelli
- Medical Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy.
| | - Alessandro Inno
- Medical Oncology Unit, Ospedale Sacro Cuore Don Calabria Cancer Care Center, Via Don A. Sempreboni 5, 37024, Negrar, VR, Italy
| | - Antonio Ghidini
- Medical Oncology Unit, Casa di Cura Igea, Via Marcona 69, 20144, Milano, Italy
| | - Lorenza Rimassa
- Medical Oncology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milano, Italy
| | - Gianluca Tomasello
- Medical Oncology Unit, ASST Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Roberto Labianca
- Medical Oncology Unit, ASST Papa Giovanni XXIII Hospital, Piazza Organizzazione Mondiale della Sanità 1, 24127, Bergamo, Italy
| | - Sandro Barni
- Medical Oncology Unit, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047, Treviglio, BG, Italy
| | | |
Collapse
|
25
|
Abstract
Pancreatic cancer remains one of the most lethal cancers. These patients often have multiple symptoms, and integrated supportive care is critical in helping them remain well for as long as possible. Fluorouracil-based chemotherapy is known to improve overall survival (OS) by approximately 3 months, compared to the best supportive care alone. A 1997 study comparing gemcitabine and fluorouracil treatment of advanced pancreatic cancer patients showed an improvement in OS of 1 month in patients receiving gemcitabine. Over the next 10 years, multiple randomized studies compared single-agent gemcitabine with combination chemotherapy and showed no effective survival improvement. However, the addition of erlotinib, an epidermal growth factor receptor (EGFR) inhibitor, was associated with a significant improvement in OS of approximately 2 weeks. However, adoption of this regimen has not been widespread because of its limited effect and added toxicity. Two clinical trials have recently prolonged OS in advanced pancreatic cancer patients by almost 1 year. The first compared FOLFIRINOX with gemcitabine alone, and was associated with a significant improvement in median survival. The second compared gemcitabine and nab-paclitaxel with gemcitabine alone, and was associated with improvements in OS. At present, these regimens are considered standard treatment for patients with good performance statuses.
Collapse
Affiliation(s)
- Hee Seung Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Woo Park
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Sarabi M, Mais L, Oussaid N, Desseigne F, Guibert P, De La Fouchardiere C. Use of gemcitabine as a second-line treatment following chemotherapy with folfirinox for metastatic pancreatic adenocarcinoma. Oncol Lett 2017; 13:4917-4924. [PMID: 28599496 DOI: 10.3892/ol.2017.6061] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/31/2017] [Indexed: 12/11/2022] Open
Abstract
There is a lack of prospective data about second-line treatments for metastatic pancreatic ductal adenocarcinoma patients. This is partially due to recent changes in first-line chemotherapy treatments. Despite this dearth of information, 50.0% of the patients who experience failure with first-line folinic acid, 5-fluorouracil, irinotecan and oxaliplatin (folfirinox) treatment are eligible for additional chemotherapy. In this setting, gemcitabine is widely used without any standard recommendations available. The present study evaluated 42 patients who received gemcitabine subsequent to a first-line treatment of folfirinox between January 2008 and December 2012 at the Centre Léon Bérard (Lyon, France). Clinical data, biological data and tumor characteristics were retrospectively analyzed to identify prognostic factors for successful treatment with gemcitabine. In total, 11 patients (26.2%) experienced control of their cancer with gemcitabine treatment. However, there was no predictive marker for their response to the drug. The median overall survival was 3.6 months from gemcitabine initiation [95% confidence interval (CI), 2.1-5.1]. The median length of gemcitabine treatment was 1.5 months (95% CI, 0.3-13.3). Among the 11 patients who were successfully treated with gemcitabine, 6 were resistant to first-line folfirinox treatment. Patients who were non responsive to folfirinox had a higher probability of success with gemcitabine compared with patients that responded to folfirinox (54.5 vs. 21.4%, respectively; P=0.061). The present study did not identify any clinical or biological marker with a predictive value for successful gemcitabine treatment. Furthermore, successful gemcitabine treatment was not correlated with patients' response to first-line folfirinox treatment. This suggests an absence of cross-resistance in the chemotherapy protocols and provides evidence for effective cancer treatment with the second-line gemcitabine therapy.
Collapse
Affiliation(s)
- Matthieu Sarabi
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Laetitia Mais
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Nadia Oussaid
- Department of Biostatistics, Centre Léon Bérard, 69008 Lyon, France
| | | | - Pierre Guibert
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | | |
Collapse
|
27
|
Second-line chemotherapy for advanced pancreatic cancer: Which is the best option? Crit Rev Oncol Hematol 2017; 115:1-12. [PMID: 28602164 DOI: 10.1016/j.critrevonc.2017.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 02/28/2017] [Accepted: 03/21/2017] [Indexed: 12/17/2022] Open
Abstract
Despite recent biological insight and therapeutic advances, the prognosis of advanced pancreatic cancer still remains poor. For more than 15 years, gemcitabine monotherapy has been the cornerstone of first-line treatment. Recently, prospective randomized trials have shown that novel upfront combination regimens tested in prospective randomized trials have resulted in improved patients' outcome increasing the proportion of putative candidate to second-line therapy. There is no definite standard of care after disease progression. A novel formulation in which irinotecan is encapsulated into liposomal-based nanoparticles may increase the efficacy of the drug without incrementing its toxicity. NAPOLI-1 was the first randomized trial to compare nanoliposomal irinotecan and fluorouracil-leucovorin (5-FU/LV) to 5-FU/LV alone after a gemcitabine-based chemotherapy. This review focuses on the current data for the management of second-line treatment for metastatic pancreatic adenocarcinoma, presents the most interesting ongoing clinical trials and illustrates the biologically-driven future options beyond disease progression.
Collapse
|
28
|
Kipps E, Young K, Starling N. Liposomal irinotecan in gemcitabine-refractory metastatic pancreatic cancer: efficacy, safety and place in therapy. Ther Adv Med Oncol 2017; 9:159-170. [PMID: 28344661 PMCID: PMC5349428 DOI: 10.1177/1758834016688816] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease. The majority of patients are diagnosed with locally advanced or metastatic disease with a prognosis of short months. Therapeutic options are limited and until recently, there was no standard second-line chemotherapy option. Liposomal constructs have been engineered to encapsulate chemotherapy thereby preventing premature metabolism, improving distribution and minimizing toxicity. Favourable preclinical data on liposomal irinotecan and early phase trials, led to a recently published phase III trial of liposomal irinotecan in combination with fluorouracil and folinic acid in patients with metastatic PDAC, who progressed after gemcitabine-based chemotherapy. As a direct result, the United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) have approved the use of liposomal irinotecan in this setting. However, first-line treatment options for this disease now include the combination regimen, FOLFIRINOX, in patients with good performance status, and the role of second-line combination treatment with liposomal irinotecan in this setting is unclear. Recent advances have changed the therapeutic landscape, as clinicians are now able to choose a sequential approach to treatment tailored to the individual patient characteristics. This article reviews current treatment options for metastatic PDAC and focuses on the efficacy, safety and place in therapy of liposomal irinotecan.
Collapse
Affiliation(s)
- Emma Kipps
- Royal Marsden NHS Foundation Trust Ringgold standard institution - Department of Gastrointestinal Oncology, London, UK
| | - Kate Young
- Royal Marsden NHS Foundation Trust Ringgold standard institution - Department of Gastrointestinal Oncology, London, UK
| | - Naureen Starling
- Royal Marsden NHS Foundation Trust Ringgold standard institution - Department of Gastrointestinal Oncology, London, UK
| |
Collapse
|
29
|
|
30
|
Ghosn M, Kourie HR, El Rassy E, Haddad FG, Hanna C, El Karak F, Nasr D. Where does chemotherapy stands in the treatment of ampullary carcinoma? A review of literature. World J Gastrointest Oncol 2016; 8:745-750. [PMID: 27795814 PMCID: PMC5064052 DOI: 10.4251/wjgo.v8.i10.745] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 07/18/2016] [Accepted: 08/18/2016] [Indexed: 02/05/2023] Open
Abstract
Ampullary carcinoma (AC) is a rare gastrointestinal tumor without clear treatment recommendations. The management of this tumor is usually extrapolated from the treatment of pancreatic, biliary duct and intestinal cancers. Few papers have studied the AC as an independent entity and yet succombs to several limitations. These studies were retrospective single institutional experiences with limited sample sizes recruited over a long period of time. Unlike metastatic ACs where chemotherapy is the only recommended option, localized AC once excised may be approached by either chemotherapy alone or concomitant chemoradiation therapy. In this review, we report the overall survival and recurrence factors of more than 1000 patients from all the studies treating exclusively ACs. We also review the medical treatment of this tumor and conclude to the necessity of multi-institutional randomized controlled studies for AC exclusively.
Collapse
|
31
|
Yang X, Xu Y, Brooks A, Guo B, Miskimins KW, Qian SY. Knockdown delta-5-desaturase promotes the formation of a novel free radical byproduct from COX-catalyzed ω-6 peroxidation to induce apoptosis and sensitize pancreatic cancer cells to chemotherapy drugs. Free Radic Biol Med 2016; 97:342-350. [PMID: 27368132 PMCID: PMC5807006 DOI: 10.1016/j.freeradbiomed.2016.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 11/26/2022]
Abstract
Recent research has demonstrated that colon cancer cell proliferation can be suppressed in the cells that overexpress COX-2 via generating 8-hydroxyoctanoic acid (a free radical byproduct) during dihomo-γ-linolenic acid (DGLA, an ω-6 fatty acid) peroxidation from knocking down cellular delta-5-desaturase (D5D, the key enzyme for converting DGLA to the downstream ω-6, arachidonic acid). Here, this novel research finding is extended to pancreatic cancer growth, as COX-2 is also commonly overexpressed in pancreatic cancer. The pancreatic cancer cell line, BxPC-3 (with high COX-2 expression and mutated p53), was used to assess not only the inhibitory effects of the enhanced formation of 8-hydroxyoctanoic acid from cellular COX-2-catalyzed DGLA peroxidation but also its potential synergistic and/or additive effect on current chemotherapy drugs. This work demonstrated that, by inducing DNA damage through inhibition of histone deacetylase, a threshold level of 8-hydroxyoctanoic acid achieved in DGLA-treated and D5D-knockdown BxPC-3 cells subsequently induce cancer cell apoptosis. Furthermore, it was shown that a combination of D5D knockdown along with DGLA treatment could also significantly sensitize BxPC-3 cells to various chemotherapy drugs, likely via a p53-independent pathway through downregulating of anti-apoptotic proteins (e.g., Bcl-2) and activating pro-apoptotic proteins (e.g., caspase 3, -9). This study reinforces the supposition that using commonly overexpressed COX-2 for molecular targeting, a strategy conceptually distinct from the prevailing COX-2 inhibition strategy used in cancer treatment, is an important as well as viable alternative to inhibit cancer cell growth. Based on the COX-2 metabolic cascade, the outcomes presented here could guide the development of a novel ω-6-based dietary care strategy in combination with chemotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Yi Xu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Amanda Brooks
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Bin Guo
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Keith W Miskimins
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, United States
| | - Steven Y Qian
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States.
| |
Collapse
|
32
|
Renouf DJ, Hedley D, Krzyzanowska MK, Schmuck M, Wang L, Moore MJ. A phase II study of the HSP90 inhibitor AUY922 in chemotherapy refractory advanced pancreatic cancer. Cancer Chemother Pharmacol 2016; 78:541-5. [PMID: 27422303 DOI: 10.1007/s00280-016-3102-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVES AUY922 is a novel heat shock protein inhibitor with preclinical activity in pancreatic cancer. This phase II study evaluated the efficacy of AUY922 in patients with advanced pancreatic cancer previously treated with chemotherapy. METHODS In this single-arm, Simon two-stage phase II trial, patients with metastatic or locally advanced pancreatic ductal adenocarcinoma who had progressed on at least one line of chemotherapy and were of good performances status (ECOG 0 or 1) were treated with AUY922 at a dose of 70 mg/m(2) IV weekly. The primary endpoint was disease control rate (objective response and stable disease ≥16 weeks). RESULTS Twelve patients were accrued, all of whom received treatment. At least possibly related ≥grade 3 adverse events included fatigue (8 %) and AST elevation (8 %). Ten patients were evaluable for response with 1 (10 %) having stable disease and 9 (90 %) progressive disease. The median progression-free survival was 1.6 months, and the median overall survival was 2.9 months. CONCLUSIONS AUY922 was not associated with significant efficacy in previously treated patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- D J Renouf
- British Columbia Cancer Agency, University of British Columbia, 600 West 10th Avenue, Vancouver, BC, V5Z4E6, Canada.
| | - D Hedley
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - M K Krzyzanowska
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - M Schmuck
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - L Wang
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - M J Moore
- British Columbia Cancer Agency, University of British Columbia, 600 West 10th Avenue, Vancouver, BC, V5Z4E6, Canada
| |
Collapse
|
33
|
Fotopoulos G, Syrigos K, Saif MW. Genetic factors affecting patient responses to pancreatic cancer treatment. Ann Gastroenterol 2016; 29:466-476. [PMID: 27708512 PMCID: PMC5049553 DOI: 10.20524/aog.2016.0056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/05/2016] [Indexed: 12/15/2022] Open
Abstract
Cancer of the exocrine pancreas is a malignancy with a high lethal rate. Surgical resection is the only possible curative mode of treatment. Metastatic pancreatic cancer is incurable with modest results from the current treatment options. New genomic information could prove treatment efficacy. An independent review of PubMed and ScienceDirect databases was performed up to March 2016, using combinations of terms such pancreatic exocrine cancer, chemotherapy, genomic profile, pancreatic cancer pharmacogenomics, genomics, molecular pancreatic pathogenesis, and targeted therapy. Recent genetic studies have identified new markers and therapeutic targets. Our current knowledge of pancreatic cancer genetics must be further advanced to elucidate the molecular basis and pathogenesis of the disease, improve the accuracy of diagnosis, and guide tailor-made therapies.
Collapse
Affiliation(s)
- George Fotopoulos
- Oncology Unit, Third Department of Medicine, University of Athens, Athens, Greece (George Fotopoulos, Konstantinos Syrigos)
| | - Konstantinos Syrigos
- Oncology Unit, Third Department of Medicine, University of Athens, Athens, Greece (George Fotopoulos, Konstantinos Syrigos); Yale School of Medicine, New Haven, CT, USA (Konstantinos Syrigos)
| | - Muhammad Wasif Saif
- Tufts University School of Medicine, Boston, Massachusetts, USA (Muhammad Wasif Saif)
| |
Collapse
|
34
|
Johnson B, Vanderwalde A, Javadi N, Feldman R, Reddy SB. Molecular profiling of a case of advanced pancreatic cancer identifies an active and tolerable combination of targeted therapy with backbone chemotherapy. J Gastrointest Oncol 2016; 7:E6-E12. [PMID: 27034805 PMCID: PMC4783749 DOI: 10.3978/j.issn.2078-6891.2015.091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/01/2015] [Indexed: 12/11/2022] Open
Abstract
Typical survival with common 1(st)-line regimens for pancreatic cancer range from 6-11 months. We report a case of a patient with stage IVB pancreatic adenocarcinoma treated with gemcitabine and erlotinib who stopped therapy after 3 months without achieving a response due to intolerance. To decide upon additional treatment options, molecular analysis was performed on liver metastasis which revealed KRAS, FBXW7, APC, and ATM mutations, with thymidylate synthase (TS) negativity and PD-1 positivity. Based on this profile of TS negativity and ATM mutation, a combination strategy was devised consisting of capecitabine, oxaliplatin, bevacizumab and vorinostat. The patient had a near complete response to therapy with this regimen. In refractory metastatic pancreatic cancer, responses of this magnitude are rarely seen. To our knowledge, this represents the first demonstrated activity of this combination in the metastatic setting which could prompt further investigation of its use in large scale clinical trials.
Collapse
|
35
|
Spadi R, Brusa F, Ponzetti A, Chiappino I, Birocco N, Ciuffreda L, Satolli MA. Current therapeutic strategies for advanced pancreatic cancer: A review for clinicians. World J Clin Oncol 2016; 7:27-43. [PMID: 26862489 PMCID: PMC4734936 DOI: 10.5306/wjco.v7.i1.27] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/22/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) would become the second leading cause of cancer death in the near future, despite representing only 3% of new cancer diagnosis. Survival improvement will come from a better knowledge of risk factors, earlier diagnosis, better integration of locoregional and systemic therapies, as well as the development of more efficacious drugs rising from a deeper understanding of disease biology. For patients with unresectable, non-metastatic disease, combined strategies encompassing primary chemotherapy and radiation seems to be promising. In fit patients, new polychemotherapy regimens can lead to better outcomes in terms of slight but significant survival improvement associated with a positive impact on quality of life. The upfront use of these regimes can also increase the rate of radical resections in borderline resectable and locally advanced PC. Second line treatments showed to positively affect both overall survival and quality of life in fit patients affected by metastatic disease. At present, oxaliplatin-based regimens are the most extensively studied. Nonetheless, other promising drugs are currently under evaluation. Presently, in addition to surgery and conventional radiation therapy, new locoregional treatment techniques are emerging as alternative options in the multimodal approach to patients or diseases not suitable for radical surgery. As of today, in contrast with other types of cancer, targeted therapies failed to show relevant activity either alone or in combination with chemotherapy and, thus, current clinical practice does not include them. Up to now, despite the fact of extremely promising results in different tumors, also immunotherapy is not in the actual therapeutic armamentarium for PC. In the present paper, we provide a comprehensive review of the current state of the art of clinical practice and research in PC aiming to offer a guide for clinicians on the most relevant topics in the management of this disease.
Collapse
|
36
|
Ettrich TJ, Perkhofer L, von Wichert G, Gress TM, Michl P, Hebart HF, Büchner-Steudel P, Geissler M, Muche R, Danner B, Kächele V, Berger AW, Güthle M, Seufferlein T. DocOx (AIO-PK0106): a phase II trial of docetaxel and oxaliplatin as a second line systemic therapy in patients with advanced pancreatic ductal adenocarcinoma. BMC Cancer 2016; 16:21. [PMID: 26772812 PMCID: PMC4714522 DOI: 10.1186/s12885-016-2052-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/06/2016] [Indexed: 12/18/2022] Open
Abstract
Background The current study was conducted to examine the activity of a docetaxel/oxaliplatin (DocOx) combination as second line treatment for advanced pancreatic ductal adenocarcinoma (Trial registration: NCT00690300. Registered June 2, 2008) Methods DocOx is a prospective, multi-center, single arm, phase II trial using docetaxel (75 mg/m2, 60 min, d 1) and oxaliplatin (80 mg/m2, 120 min, d 2) in 21-day cycles. The treatment period was scheduled for up to 8 cycles. Primary endpoint was tumor response according to RECIST 1.0. Secondary endpoints were progression free survival, overall survival, safety/toxicity, quality of life and clinical benefit. Results Data represent the intention to treat analysis of 44 patients with chemorefractory pancreatic cancer enrolled between 2008 and 2012 at five institutions in Germany. The primary endpoint of tumor response was achieved in 15.9 % of the patients (7 partial remissions, no complete remission), with a disease control rate of 48 % after the first two treatment cycles. Median progression free survival (PFS) was 1.82 months (CI 95 % 1.5–3.96 months) and median overall survival (OS) was 10.1 months (CI 95 % 5.1–14.1 months). Conclusions This single-arm trial demonstrates that the combination of docetaxel and oxaliplatin yields promising results for the treatment of advanced pancreatic ductal adenocarcinoma patients. Selected patients had particular benefit from this treatment as indicated by long PFS and OS times. Even after 8 cycles of treatment with DocOx a partial response was observed in 2 patients and stable disease was observed in another 6 patients. The data obtained with the DocOx protocol compare well with other second line protocols such as OFF (oxaliplatin, 5-FU, leucovorin). The DocOx regimen could be an interesting option for patients who received gemcitabine as first line treatment for metastatic pancreatic cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2052-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Goetz von Wichert
- Department of Internal Medicine, Schön-Klinik Hamburg-Eilbeck, Hamburg, Germany.
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, Philipps University of Marburg, Marburg, Germany.
| | - Patrick Michl
- Department of Internal Medicine I, Martin-Luther-University, Halle (Saale), Germany.
| | - Holger F Hebart
- Department of Internal Medicine, Stauferklinikum Schwaebisch-Gmuend, Mutlangen, Germany.
| | - Petra Büchner-Steudel
- Department of Internal Medicine I, Martin-Luther-University, Halle (Saale), Germany.
| | - Michael Geissler
- Department of Internal Medicine, Oncology/Hematology, Gastroenterology, Esslingen Hospital, Esslingen, Germany.
| | - Rainer Muche
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany.
| | - Bettina Danner
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany.
| | | | - Andreas W Berger
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany.
| | - Melanie Güthle
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany.
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany.
| |
Collapse
|
37
|
Nagrial AM, Chin VT, Sjoquist KM, Pajic M, Horvath LG, Biankin AV, Yip D. Second-line treatment in inoperable pancreatic adenocarcinoma: A systematic review and synthesis of all clinical trials. Crit Rev Oncol Hematol 2015; 96:483-97. [PMID: 26481952 DOI: 10.1016/j.critrevonc.2015.07.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 05/23/2015] [Accepted: 07/16/2015] [Indexed: 12/14/2022] Open
Abstract
There remains uncertainty regarding the optimal second-line chemotherapy in advanced pancreatic ductal adenocarcinoma (PDAC). The current recommendation of 5-fluorouracil and oxaliplatin may not be relevant in current practice, as FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan and oxaliplatin) has become a more popular first line therapy in fit patients. The majority of studies in this setting are single-arm Phase II trials with significant heterogeneity of patient populations, treatments and outcomes. In this review, we sought to systematically review and synthesise all prospective data available for the second-line treatment of advanced PDAC.
Collapse
Affiliation(s)
- Adnan M Nagrial
- The Kinghorn Cancer Centre, 370 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia.
| | - Venessa T Chin
- The Kinghorn Cancer Centre, 370 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Katrin M Sjoquist
- NHMRC Clinical Trials Centre, University of Sydney, NSW, Australia; Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, 370 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; St. Vincents's Clinical School, Faculty of Medicine, University of NSW, Australia
| | - Lisa G Horvath
- The Kinghorn Cancer Centre, 370 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW 2050, Australia
| | - Andrew V Biankin
- The Kinghorn Cancer Centre, 370 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, NSW 2200, Australia; South Western Sydney Clinical School, Faculty of Medicine, University of NSW, Liverpool, NSW 2170, Australia; Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, Scotland, UK; West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, Scotland G4 0SF, UK
| | - Desmond Yip
- Department of Medical Oncology, The Canberra Hospital, Garran, ACT, Australia; ANU Medical School, Australian National University, Acton, ACT, Australia
| |
Collapse
|
38
|
Nishiofuku H, Tanaka T, Marugami N, Sho M, Akahori T, Nakajima Y, Kichikawa K. Increased tumour ADC value during chemotherapy predicts improved survival in unresectable pancreatic cancer. Eur Radiol 2015; 26:1835-42. [PMID: 26385808 PMCID: PMC4863905 DOI: 10.1007/s00330-015-3999-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/25/2015] [Accepted: 09/02/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVES To investigate whether changes to the apparent diffusion coefficient (ADC) of primary tumour in the early period after starting chemotherapy can predict progression-free survival (PFS) or overall survival (OS) in patients with unresectable pancreatic adenocarcinoma. METHODS Subjects comprised 43 patients with histologically confirmed unresectable pancreatic cancer treated with first-line chemotherapy. Minimum ADC values in primary tumour were measured using the selected area ADC (sADC), which excluded cystic and necrotic areas and vessels, and the whole tumour ADC (wADC), which included whole tumour components. Relative changes in ADC were calculated from baseline to 4 weeks after initiation of chemotherapy. Relationships between ADC and both PFS and OS were modelled by Cox proportional hazards regression. RESULTS Median PFS and OS were 6.1 and 11.0 months, respectively. In multivariate analysis, sADC change was the strongest predictor of PFS (hazard ratio (HR), 4.5; 95 % confidence interval (CI), 1.7-11.9; p = 0.002). Multivariate Cox regression analysis for OS revealed sADC change and CRP as independent predictive markers, with sADC change as the strongest predictive biomarker (HR, 6.7; 95 % CI, 2.7-16.6; p = 0.001). CONCLUSION Relative changes in sADC could provide a useful imaging biomarker to predict PFS and OS with chemotherapy for unresectable pancreatic adenocarcinoma. KEY POINTS • Relative change in ADC value can predict survival in unresectable pancreatic cancer. • ADC change could determine a chemosensitivity of pancreatic cancer. • ADC values should be measured by excluding cystic, necrotic areas and vessels.
Collapse
Affiliation(s)
- Hideyuki Nishiofuku
- Department of Radiology and IVR Center, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan.
| | - Toshihiro Tanaka
- Department of Radiology and IVR Center, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan
| | - Nagaaki Marugami
- Department of Endoscopy and Ultrasound, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan
| | - Takahiro Akahori
- Department of Surgery, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan
| | - Yoshiyuki Nakajima
- Department of Surgery, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan
| | - Kimihiko Kichikawa
- Department of Radiology and IVR Center, Nara Medical University, Kashihara-city, Nara, 634-8522, Japan
| |
Collapse
|
39
|
Onesti CE, Romiti A, Roberto M, Falcone R, Marchetti P. Recent advances for the treatment of pancreatic and biliary tract cancer after first-line treatment failure. Expert Rev Anticancer Ther 2015; 15:1183-98. [PMID: 26325474 DOI: 10.1586/14737140.2015.1081816] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Here, we evaluate clinical trials on chemotherapy for patients with pancreatic or biliary tract cancer after first-line treatment failure. Clinical trials on conventional and innovative medical treatments for progressive pancreatic and biliary cancer were analyzed. Metronomic chemotherapy, which consists of the administration of continuative low-dose of anticancer drugs, was also considered. A significant extension of overall survival was achieved with second-line, regimens in patients with gemcitabine-refractory pancreatic cancer. Moreover, many Phase II studies, including chemotherapy and target molecules and immunotherapy, have reported promising results, in both pancreatic and biliary cancer. However, data in these patients' setting are very heterogeneous, and only few randomized studies are available.
Collapse
Affiliation(s)
| | | | - Michela Roberto
- a Clinical and Molecular Medicine Department, Sapienza University, Rome, Italy
| | - Rosa Falcone
- a Clinical and Molecular Medicine Department, Sapienza University, Rome, Italy
| | - Paolo Marchetti
- a Clinical and Molecular Medicine Department, Sapienza University, Rome, Italy
| |
Collapse
|
40
|
Randomised phase II trial of S-1 plus oxaliplatin vs S-1 in patients with gemcitabine-refractory pancreatic cancer. Br J Cancer 2015; 112:1428-34. [PMID: 25880004 PMCID: PMC4453667 DOI: 10.1038/bjc.2015.103] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/07/2015] [Accepted: 02/23/2015] [Indexed: 12/17/2022] Open
Abstract
Background: This randomised, open-label, multicenter phase II study compared progression-free survival (PFS) of S-1 plus oxaliplatin (SOX) with that of S-1 alone in patients with gemcitabine-refractory pancreatic cancer. Methods: Patients with confirmed progressive disease following the first-line treatment with a gemcitabine-based regimen were randomised to receive either S-1 (80/100/120 mg day−1 based on body surface area (BSA), orally, days 1–28, every 6 weeks) or SOX (S-1 80/100/120 mg day−1 based on BSA, orally, days 1–14, plus oxaliplatin 100 mg m−2, intravenously, day 1, every 3 weeks). The primary end point was PFS. Results: Between January 2009 and July 2010, 271 patients were randomly allocated to either S-1 (n=135) or SOX (n=136). Median PFS for S-1 and SOX were 2.8 and 3.0 months, respectively (hazard ratio (HR)=0.84; 95% confidence interval (CI), 0.65–1.08; stratified log-rank test P=0.18). Median overall survival (OS) was 6.9 vs 7.4 months (HR=1.03; 95% CI, 0.79–1.34; stratified log-rank test P=0.82). The response rate (RR) was 11.5% vs 20.9% (P=0.04). The major grade 3/4 toxicities (S-1 and SOX) were neutropenia (11.4% and 8.1%), thrombocytopenia (4.5% and 10.3%) and anorexia (12.9% and 14.7%). Conclusions: Although SOX showed an advantage in RR, it provided no significant improvement in PFS or OS compared with S-1 alone.
Collapse
|
41
|
Sclafani F, Iyer R, Cunningham D, Starling N. Management of metastatic pancreatic cancer: Current treatment options and potential new therapeutic targets. Crit Rev Oncol Hematol 2015; 95:318-36. [PMID: 25921418 DOI: 10.1016/j.critrevonc.2015.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/18/2015] [Accepted: 03/31/2015] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a malignancy with a poor prognosis, with the majority of patients diagnosed with advanced disease on presentation. Treatment options remain limited with little progress over the last 40 years. This review will focus on the current management of metastatic pancreatic ductal adenocarcinoma, with a discussion of new and future treatment strategies based on an improved understanding of tumour biology and mechanisms of pathogenesis.
Collapse
Affiliation(s)
| | - Ridhima Iyer
- The Royal Marsden NHS Foundation Trust, London and Surrey, UK
| | | | | |
Collapse
|
42
|
Phua VCE, Wong WQ, Tan PL, Bustam AZ, Saad M, Alip A, Ishak WZW. Capecitabine Pattern of Usage, Rate of Febrile Neutropaenia and Treatment Related Death in Asian Cancer Patients in Clinical Practice. Asian Pac J Cancer Prev 2015; 16:1449-53. [DOI: 10.7314/apjcp.2015.16.4.1449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
43
|
Pancreatic cancer: diagnosis and treatments. Tumour Biol 2015; 36:1375-84. [PMID: 25680410 DOI: 10.1007/s13277-015-3223-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the deadliest cancers, with exceptionally high mortality. Despite the relatively low incidence rate (10th), it is the fourth leading cause of cancer-related deaths in most developed countries. To improve the early diagnosis of pancreatic cancer and strengthen the standardized comprehensive treatment are still the main focus of pancreatic cancer research. Here, we summarized the rapid developments in the diagnosis and treatments of pancreatic cancer. Regarding diagnosis, we reviewed advances in medical imaging technology, tumor markers, molecular biology (e.g., gene mutation), and proteomics. Moreover, great progress has also been made in the treatments of this disease, including surgical resection, chemotherapy, targeted radiotherapy, targeted minimally invasive treatment, and molecular targeted therapy. Therefore, we also recapitulated the development, advantages, and disadvantages of each of the treatment methods in this review.
Collapse
|
44
|
Maier-Stocker C, Bitzer M, Malek NP, Plentz RR. Analysis of second-line chemotherapies for ductal pancreatic adenocarcinoma in a German single-center cohort. Scand J Gastroenterol 2014; 49:1480-5. [PMID: 25390691 DOI: 10.3109/00365521.2014.978816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third most common tumor of the gastrointestinal tract. At the time of diagnosis, the majority of PDACs shows already metastasis and does not qualify for curative surgery. Therefore, palliative chemotherapy has a very high priority, but recommendations after failure of first-line chemotherapies are quite limited. The aim of our analysis was to evaluate the efficacy of different second-line treatments after pretreatment with gemcitabine (57.5%), gemcitabine + erlotinib (25%), and platinum-based chemotherapy (17.5%). We included all patients with advanced PDAC treated with second-line chemotherapy in our department between 2005 and 2012. A total of 22 patients were treated with XELOX, 8 patients with FOLFOX, 6 patients with gemcitabine (+/- erlotinib) and 4 patients with FOLFIRI. On average, the patients received 4.2 cycles (standard deviation [SD] SD: 3.5) over a period of 2.5 months (SD: 2.6). The median overall survival (OS) for all patients was 5.4 months, progression-free survival was 3.5 months, and a tumor control was achieved in 21% of all cases. Toxicity profile was acceptable between the second-line chemotherapies and there was no significant difference in the other investigated end points. Interestingly, there was also no effect of the first-line treatment and their duration for the OS of the second-line therapy. According to our findings, second-line chemotherapies in advanced PDAC are beneficial and should be offered to patients, but we did not detect any superiority of a specific drug combination. More prospective, randomized and larger studies are necessary to evaluate new strategies for second-line chemotherapies.
Collapse
|
45
|
Bayoglu IV, Varol U, Yildiz I, Muslu U, Alacacioglu A, Kucukzeybek Y, Akyol M, Demir L, Dirican A, Cokmert S, Yildiz Y, Karabulut B, Uslu R, Tarhan MO. Second-Line Capecitabine and Oxaliplatin Combination for Gemcitabine-Resistant Advanced Pancreatic Cancer. Asian Pac J Cancer Prev 2014; 15:7119-23. [DOI: 10.7314/apjcp.2014.15.17.7119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
46
|
Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, Görner M, Mölle M, Greten TF, Lakner V, Bischoff S, Sinn M, Dörken B, Pelzer U. Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: outcomes from the CONKO-003 trial. J Clin Oncol 2014; 32:2423-9. [PMID: 24982456 DOI: 10.1200/jco.2013.53.6995] [Citation(s) in RCA: 324] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To assess the efficacy of a second-line regimen of oxaliplatin and folinic acid-modulated fluorouracil in patients with advanced pancreatic cancer who have experienced progression while receiving gemcitabine monotherapy. PATIENTS AND METHODS A randomized, open-label, phase III study was conducted in 16 institutions throughout Germany. Recruitment ran from January 2004 until May 2007, and the last follow-up concluded in December 2012. Overall, 168 patients age 18 years or older who experienced disease progression during first-line gemcitabine therapy were randomly assigned to folinic acid and fluorouracil (FF) or oxaliplatin and FF (OFF). Patients were stratified according to the presence of metastases, duration of first-line therapy, and Karnofsky performance status. RESULTS Median follow-up was 54.1 months, and 160 patients were eligible for the primary analysis. The median overall survival in the OFF group (5.9 months; 95% CI, 4.1 to 7.4) versus the FF group (3.3 months; 95% CI, 2.7 to 4.0) was significantly improved (hazard ratio [HR], 0.66; 95% CI, 0.48 to 0.91; log-rank P = .010). Time to progression with OFF (2.9 months; 95% CI, 2.4 to 3.2) versus FF (2.0 months; 95% CI, 1.6 to 2.3) was significantly extended also (HR, 0.68; 95% CI, 0.50 to 0.94; log-rank P = .019). Rates of adverse events were similar between treatment arms, with the exception of grades 1 to 2 neurotoxicity, which were reported in 29 patients (38.2%) and six patients (7.1%) in the OFF and FF groups, respectively (P < .001). CONCLUSION Second-line OFF significantly extended the duration of overall survival when compared with FF alone in patients with advanced gemcitabine-refractory pancreatic cancer.
Collapse
Affiliation(s)
- Helmut Oettle
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany.
| | - Hanno Riess
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Jens M Stieler
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Gerhard Heil
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Ingo Schwaner
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Jörg Seraphin
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Martin Görner
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Matthias Mölle
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Tim F Greten
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Volker Lakner
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Sven Bischoff
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Marianne Sinn
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Bernd Dörken
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| | - Uwe Pelzer
- Helmut Oettle, Hanno Riess, Jens M. Stieler, Sven Bischoff, Marianne Sinn, Bernd Dörken, and Uwe Pelzer, Charitě Universitätsmedizin; Ingo Schwaner, Clinical Center, Berlin; Helmut Oettle, Clinical Center, Friedrichschafen; Gerhard Heil, Clinical Center, Lüdenscheid; Jörg Seraphin, Clinical Center, Northeim; Martin Görner, Clinical Center, Bielefeld; Matthias Mölle, Clinical Center, Dresden; Tim F. Greten, Hannover Medical School, Hannover; and Volker Lakner, Clinical Center, Rostock, Germany
| |
Collapse
|
47
|
Self-assembled nanoscale coordination polymers with trigger release properties for effective anticancer therapy. Nat Commun 2014; 5:4182. [PMID: 24964370 PMCID: PMC4181838 DOI: 10.1038/ncomms5182] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 05/21/2014] [Indexed: 12/23/2022] Open
Abstract
Nanoscale coordination polymers (NCPs) are self-assembled from metal ions and organic bridging ligands, and can overcome many drawbacks of existing drug delivery systems by virtue of tunable compositions, sizes, and shapes; high drug loadings; ease of surface modification; and intrinsic biodegradability. Here we report the self-assembly of zinc bisphosphonate NCPs that carry 48±3 wt% cisplatin prodrug and 45±5 wt% oxaliplatin prodrug. In vivo pharmacokinetic studies in mice show minimal uptake of pegylated NCPs by the mononuclear phagocyte system and excellent blood circulation half-lives of 16.4±2.9 and 12.0±3.9 h for the NCPs carrying cisplatin and oxaliplatin, respectively. In all tumor xenograft models evaluated, including CT26 colon cancer, H460 lung cancer, and AsPC-1 pancreatic cancer, pegylated NCPs show superior potency and efficacy compared to free drugs. As the first example of using NCPs as nanotherapeutics with enhanced antitumor activities, this study establishes NCPs as a promising drug delivery platform for cancer therapy.
Collapse
|
48
|
|
49
|
Zaanan A, Trouilloud I, Markoutsaki T, Gauthier M, Dupont-Gossart AC, Lecomte T, Aparicio T, Artru P, Thirot-Bidault A, Joubert F, Fanica D, Taieb J. FOLFOX as second-line chemotherapy in patients with pretreated metastatic pancreatic cancer from the FIRGEM study. BMC Cancer 2014; 14:441. [PMID: 24929865 PMCID: PMC4075567 DOI: 10.1186/1471-2407-14-441] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 06/09/2014] [Indexed: 02/06/2023] Open
Abstract
Background FOLFOX second-line treatment seems to be a validated option for patients with pancreatic cancer (PC) progressing after gemcitabine chemotherapy. However, other therapeutics strategy has developed in first-line therapy, as the FIRGEM phase II study that evaluated gemcitabine alone versus FOLFIRI.3 alternating with gemcitabine every two months. The present study assessed the efficacy and safety of FOLFOX after failure of the first-line therapy used in the FIRGEM study. Methods In this prospective observational cohort study, we analysed all consecutive patients who received second-line chemotherapy with FOLFOX among 98 patients with metastatic PC included in the FIRGEM study. Progression-free survival (PFS) and overall survival (OS) were estimated from the start of second-line chemotherapy using the Kaplan-Meier method. Results Among 46 patients who received second-line chemotherapy, 27 patients (male, 55%; median age, 61 years; performance status (PS) 0–1, 44%) were treated with FOLFOX after progression to first-line gemcitabine alone (n = 20) or FOLFIRI.3 alternating with gemcitabine (n = 7). Grade 3 toxicity was observed in 33% of patients (no grade 4 toxicity). At the end of follow-up, all patients had progressed and 25 had died. No objective response was observed, and disease control rate was 36%. Median PFS and OS were 1.7 and 4.3 months, respectively. In multivariate analysis, PS was the only independent prognostic factor. For patients PS 0–1 versus 2–3, median PFS was 3.0 versus 1.2 months (log rank, p = 0.002), and median OS was 5.9 versus 2.6 months (log rank, p = 0.001). Conclusions This study suggests that FOLFOX second-line therapy offered interesting efficacy results with an acceptable toxicity profile in metastatic PC patients with a good PS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, AP-HP, 20 rue Leblanc, 75015 Paris, France.
| |
Collapse
|
50
|
Renouf DJ, Tang PA, Hedley D, Chen E, Kamel-Reid S, Tsao MS, Tran-Thanh D, Gill S, Dhani N, Au HJ, Wang L, Moore MJ. A phase II study of erlotinib in gemcitabine refractory advanced pancreatic cancer. Eur J Cancer 2014; 50:1909-15. [PMID: 24857345 DOI: 10.1016/j.ejca.2014.04.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/21/2014] [Accepted: 04/02/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Erlotinib induced skin toxicity has been associated with clinical benefit in several tumour types. This phase II study evaluated the efficacy of erlotinib, dose escalated to rash, in patients with advanced pancreatic cancer previously treated with gemcitabine. METHODS Erlotinib was given at an initial dose of 150 mg/day, and the dose was escalated by 50mg every 2 weeks (to a maximum of 300 mg/day) until >grade 1 rash or other dose limiting toxicities occurred. Erlotinib pharmacokinetics were performed, and baseline tumour tissue was collected for mutational analysis and epidermal growth factor receptor (EGFR) expression. The primary end-point was the disease control rate (objective response and stable disease >8 weeks). RESULTS Fifty-one patients were accrued, and 49 received treatment. Dose-escalation to 200-300 mg of erlotinib was possible in 9/49 (18%) patients. The most common ⩾ grade 3 adverse events included fatigue (6%), rash (4%) and diarrhoea (4%). Thirty-seven patients were evaluable for response, and the best response was stable disease in 12 patients (32% (95% confidence interval (CI) 17-47%)). Disease control was observed in nine patients (24% (95% CI: 10-38%)). Median survival was 3.8 months, and 6 month overall survival rate was 32% (95% CI 19-47%). Mutational analysis and EGFR expression were performed on 29 patients, with 93% having KRAS mutations, none having EGFR mutations, and 86% expressing EGFR. Neither KRAS mutational status nor EGFR expression was associated with survival. CONCLUSIONS Erlotinib dose escalated to rash was well tolerated but not associated with significant efficacy in non-selected patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- D J Renouf
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada.
| | - P A Tang
- Tom Baker Cancer Centre, Calgary, AB, Canada
| | - D Hedley
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - E Chen
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - S Kamel-Reid
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - M S Tsao
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - D Tran-Thanh
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - S Gill
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - N Dhani
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - H J Au
- Cross Cancer Institute, Edmonton, AB, Canada
| | - L Wang
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - M J Moore
- University Health Network-Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|