751
|
Obaid G, Broekgaarden M, Bulin AL, Huang HC, Kuriakose J, Liu J, Hasan T. Photonanomedicine: a convergence of photodynamic therapy and nanotechnology. NANOSCALE 2016; 8:12471-503. [PMID: 27328309 PMCID: PMC4956486 DOI: 10.1039/c5nr08691d] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
As clinical nanomedicine has emerged over the past two decades, phototherapeutic advancements using nanotechnology have also evolved and impacted disease management. Because of unique features attributable to the light activation process of molecules, photonanomedicine (PNM) holds significant promise as a personalized, image-guided therapeutic approach for cancer and non-cancer pathologies. The convergence of advanced photochemical therapies such as photodynamic therapy (PDT) and imaging modalities with sophisticated nanotechnologies is enabling the ongoing evolution of fundamental PNM formulations, such as Visudyne®, into progressive forward-looking platforms that integrate theranostics (therapeutics and diagnostics), molecular selectivity, the spatiotemporally controlled release of synergistic therapeutics, along with regulated, sustained drug dosing. Considering that the envisioned goal of these integrated platforms is proving to be realistic, this review will discuss how PNM has evolved over the years as a preclinical and clinical amalgamation of nanotechnology with PDT. The encouraging investigations that emphasize the potent synergy between photochemistry and nanotherapeutics, in addition to the growing realization of the value of these multi-faceted theranostic nanoplatforms, will assist in driving PNM formulations into mainstream oncological clinical practice as a necessary tool in the medical armamentarium.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tayyaba Hasan
- Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard-MIT Division of Health Science and Technology, Boston, Massachusetts, USA
| |
Collapse
|
752
|
Takahara N, Isayama H, Nakai Y, Ishigami H, Satoi S, Mizuno S, Kogure H, Matsubara S, Yamamoto N, Yamaguchi H, Tada M, Kitayama J, Watanabe T, Koike K. Intravenous and intraperitoneal paclitaxel with S-1 for treatment of refractory pancreatic cancer with malignant ascites. Invest New Drugs 2016; 34:636-42. [PMID: 27339809 DOI: 10.1007/s10637-016-0369-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/14/2016] [Indexed: 12/27/2022]
Abstract
UNLABELLED Objectives The aim of this study was to evaluate the safety and efficacy of intravenous and intraperitoneal paclitaxel (PTX) combined with S-1 for treatment of gemcitabine-refractory pancreatic cancer with malignant ascites. Methods After the feasibility of this regimen was first confirmed in an interim analysis in 10 patients, a total of 35 patients were enrolled between April 2011 and December 2014. PTX was administered intravenously (50 mg/m(2)) and intraperitoneally (20 mg/m(2)) on days 1 and 8, and 80 mg/m(2) S-1 was administered on days 1-14 every 3 weeks. The primary endpoint was overall survival (OS). The secondary endpoints were progression-free survival (PFS), the objective tumor response, efficacy against malignant ascites, and safety. Result In all 35 patients, the median OS and PFS were 4.8 (95 % confidence interval [CI], 2.1-5.3) months and 2.8 (95 % CI, 0.9-4.1) months, respectively. The 26 patients who were evaluable for efficacy achieved a response rate of 8 % and a disease control rate of 69 %. Malignant ascites had disappeared or decreased in 18 (69 %) patients, including complete resolution in 4 (15 %), and a negative change in cytological status was achieved in 8 (31 %) patients. The major grade 3/4 adverse events included neutropenia (34 %), anemia (31 %), nausea (9 %), and catheter-related infections (6 %). Conclusion Combination chemotherapy consisting of intravenous and intraperitoneal PTX with S-1 showed acceptable toxicity and favorable efficacy in pancreatic cancer patients with malignant ascites. ( CLINICAL TRIAL REGISTRATION NUMBER UMIN000005306).
Collapse
Affiliation(s)
- Naminatsu Takahara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | | | - Sohei Satoi
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Suguru Mizuno
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hirofumi Kogure
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Saburo Matsubara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Natsuyo Yamamoto
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hironori Yamaguchi
- Department of Surgery, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Minoru Tada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Joji Kitayama
- Department of Surgery, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Toshiaki Watanabe
- Department of Surgical Oncology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
753
|
Passero FC, Grapsa D, Syrigos KN, Saif MW. The safety and efficacy of Onivyde (irinotecan liposome injection) for the treatment of metastatic pancreatic cancer following gemcitabine-based therapy. Expert Rev Anticancer Ther 2016; 16:697-703. [PMID: 27219482 DOI: 10.1080/14737140.2016.1192471] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Patients with advanced and metastatic pancreatic cancer refractory to gemcitabine based therapy have a dismal prognosis and limited therapeutic options. Recently, the FDA approved nanoliposomal irinotecan combined with fluorouracil/folinic acid for such patients based upon results of the NAPOLI-1 study which showed this regimen compared to fluorouracil/folinic acid significantly prolonged progression free survival (3.1 vs. 1.5 months) and overall survival (6.2 vs. 4.1 months). AREAS COVERED The pharmacokinetic and pharmacogenetic characteristics of this novel formulation of irinotecan, its safety profile, and use in a clinical context for patients with pancreatic cancer are reviewed. Expert commentary: Nanoliposomal irinotecan, in combination with 5-FU/folinic acid, represents an important step forward in improving second line treatment options in patients with progression of metastatic pancreatic cancer. Furthermore, the novel drug formulation offers pharmacokinetic advantages which serve as a basis for further clinical testing in a various pancreatic cancer settings and other malignancies.
Collapse
Affiliation(s)
- Frank C Passero
- a Division of Hematology/Oncology and Experimental Therapeutics , Tufts Medical Center , Boston , MA , USA
| | - Dimitra Grapsa
- b Oncology Unit GPP , Sotiria General Hospital , Athens , Greece
| | - Kostas N Syrigos
- b Oncology Unit GPP , Sotiria General Hospital , Athens , Greece
| | - Muhammad Wasif Saif
- a Division of Hematology/Oncology and Experimental Therapeutics , Tufts Medical Center , Boston , MA , USA
| |
Collapse
|
754
|
Sohal DPS, Mangu PB, Khorana AA, Shah MA, Philip PA, O'Reilly EM, Uronis HE, Ramanathan RK, Crane CH, Engebretson A, Ruggiero JT, Copur MS, Lau M, Urba S, Laheru D. Metastatic Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34:2784-96. [PMID: 27247222 DOI: 10.1200/jco.2016.67.1412] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To provide evidence-based recommendations to oncologists and others for the treatment of patients with metastatic pancreatic cancer. METHODS American Society of Clinical Oncology convened an Expert Panel of medical oncology, radiation oncology, surgical oncology, gastroenterology, palliative care, and advocacy experts to conduct a systematic review of the literature from April 2004 to June 2015. Outcomes were overall survival, disease-free survival, progression-free survival, and adverse events. RESULTS Twenty-four randomized controlled trials met the systematic review criteria. RECOMMENDATIONS A multiphase computed tomography scan of the chest, abdomen, and pelvis should be performed. Baseline performance status and comorbidity profile should be evaluated. Goals of care, patient preferences, treatment response, psychological status, support systems, and symptom burden should guide decisions for treatments. A palliative care referral should occur at first visit. FOLFIRINOX (leucovorin, fluorouracil, irinotecan, and oxaliplatin; favorable comorbidity profile) or gemcitabine plus nanoparticle albumin-bound (NAB) -paclitaxel (adequate comorbidity profile) should be offered to patients with Eastern Cooperative Oncology Group performance status (ECOG PS) 0 to 1 based on patient preference and support system available. Gemcitabine alone is recommended for patients with ECOG PS 2 or with a comorbidity profile that precludes other regimens; the addition of capecitabine or erlotinib may be offered. Patients with an ECOG PS ≥ 3 and poorly controlled comorbid conditions should be offered cancer-directed therapy only on a case-by-case basis; supportive care should be emphasized. For second-line therapy, gemcitabine plus NAB-paclitaxel should be offered to patients with first-line treatment with FOLFIRINOX, an ECOG PS 0 to 1, and a favorable comorbidity profile; fluorouracil plus oxaliplatin, irinotecan, or nanoliposomal irinotecan should be offered to patients with first-line treatment with gemcitabine plus NAB-paclitaxel, ECOG PS 0 to 1, and favorable comorbidity profile, and gemcitabine or fluorouracil should be offered to patients with either an ECOG PS 2 or a comorbidity profile that precludes other regimens. Additional information is available at www.asco.org/guidelines/MetPC and www.asco.org/guidelineswiki.
Collapse
Affiliation(s)
- Davendra P S Sohal
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Pamela B Mangu
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Alok A Khorana
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Manish A Shah
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Philip A Philip
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Eileen M O'Reilly
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Hope E Uronis
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Ramesh K Ramanathan
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Christopher H Crane
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Anitra Engebretson
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Joseph T Ruggiero
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Mehmet S Copur
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Michelle Lau
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Susan Urba
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Daniel Laheru
- Davendra P.S. Sohal and Alok A. Khorana, Cleveland Clinic, Cleveland, OH; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Manish A. Shah, The Weill Cornell Medical Center; Philip A. Philip, Karmanos Cancer Institute, Detroit; Susan Urba, University of Michigan Cancer Center, Ann Arbor, MI; Eileen M. O'Reilly, Memorial Sloan Kettering Cancer Center; Joseph T. Ruggiero, Weill Cornell Medical College, New York, NY; Hope E. Uronis, Duke University, Durham, NC; Ramesh K. Ramanathan, Mayo Clinic, Scottsdale; Michelle Lau, Community Hospital Based Cancer Center, Tempe, AZ; Christopher H. Crane, The University of Texas MD Anderson Cancer Center, Houston, TX; Anitra Engebretson, Patient Representative, Portland, OR; Mehmet S. Copur, St Francis Medical Center, Grand Island, NE; and Daniel Laheru, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
755
|
Piktel E, Niemirowicz K, Wątek M, Wollny T, Deptuła P, Bucki R. Recent insights in nanotechnology-based drugs and formulations designed for effective anti-cancer therapy. J Nanobiotechnology 2016; 14:39. [PMID: 27229857 PMCID: PMC4881065 DOI: 10.1186/s12951-016-0193-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
The rapid development of nanotechnology provides alternative approaches to overcome several limitations of conventional anti-cancer therapy. Drug targeting using functionalized nanoparticles to advance their transport to the dedicated site, became a new standard in novel anti-cancer methods. In effect, the employment of nanoparticles during design of antineoplastic drugs helps to improve pharmacokinetic properties, with subsequent development of high specific, non-toxic and biocompatible anti-cancer agents. However, the physicochemical and biological diversity of nanomaterials and a broad spectrum of unique features influencing their biological action requires continuous research to assess their activity. Among numerous nanosystems designed to eradicate cancer cells, only a limited number of them entered the clinical trials. It is anticipated that progress in development of nanotechnology-based anti-cancer materials will provide modern, individualized anti-cancer therapies assuring decrease in morbidity and mortality from cancer diseases. In this review we discussed the implication of nanomaterials in design of new drugs for effective antineoplastic therapy and describe a variety of mechanisms and challenges for selective tumor targeting. We emphasized the recent advantages in the field of nanotechnology-based strategies to fight cancer and discussed their part in effective anti-cancer therapy and successful drug delivery.
Collapse
Affiliation(s)
- Ewelina Piktel
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland
| | - Katarzyna Niemirowicz
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland
| | - Marzena Wątek
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-317, Kielce, Poland
| | - Tomasz Wollny
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-317, Kielce, Poland
| | - Piotr Deptuła
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland
| | - Robert Bucki
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2c, 15-222, Bialystok, Poland. .,Department of Physiology, Pathophysiology and Immunology of Infections, The Faculty of Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Al. IX Wieków Kielc 19, 25-317, Kielce, Poland.
| |
Collapse
|
756
|
Chiang NJ, Chang JY, Shan YS, Chen LT. Development of nanoliposomal irinotecan (nal-IRI, MM-398, PEP02) in the management of metastatic pancreatic cancer. Expert Opin Pharmacother 2016; 17:1413-20. [DOI: 10.1080/14656566.2016.1183646] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
757
|
Chen LT, Wang-Gillam A, Von Hoff DD, Bayever E, Belanger B. Nanoliposomal irinotecan in metastatic pancreatic cancer - Authors' reply. Lancet 2016; 387:1997. [PMID: 27203770 DOI: 10.1016/s0140-6736(16)30365-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.
| | | | | | | | | |
Collapse
|
758
|
Milosevic I, Drysdale H, Goldacre B. Nanoliposomal irinotecan in metastatic pancreatic cancer. Lancet 2016; 387:1997. [PMID: 27203772 DOI: 10.1016/s0140-6736(16)30463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Ioan Milosevic
- Centre for Evidence-Based Medicine, Nuffield Department of Primary Care Health Services, Oxford, OX2 6GG, UK.
| | - Henry Drysdale
- Centre for Evidence-Based Medicine, Nuffield Department of Primary Care Health Services, Oxford, OX2 6GG, UK
| | - Ben Goldacre
- Centre for Evidence-Based Medicine, Nuffield Department of Primary Care Health Services, Oxford, OX2 6GG, UK
| |
Collapse
|
759
|
Westphalen CB, Kruger S, Haas M, Heinemann V, Boeck S. Safety of palliative chemotherapy in advanced pancreatic cancer. Expert Opin Drug Saf 2016; 15:947-54. [PMID: 27070177 DOI: 10.1080/14740338.2016.1177510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pancreatic cancer is a major health burden. Currently, the majority of patients is diagnosed at advanced stages and thus qualifies for palliative chemotherapy. Gemcitabine monotherapy has been the gold standard for many years. Recently, more effective chemotherapeutic regimens have shown meaningful clinical activity in patients with metastatic pancreatic cancer. AREAS COVERED In this review we have aimed to give an overview on the treatment options for patients diagnosed with metastatic pancreatic cancer with an emphasis on the safety and toxicity of the applied regimens. We have conducted a pubmed search using the terms 'metastatic pancreatic cancer', 'palliative chemotherapy', 'safety' and 'toxicity'. Our special focus rested on randomized phase III trials to provide readers with the highest level of available evidence. EXPERT OPINION The emergence of new and more effective chemotherapy regimens gives clinicians more freedom in the treatment of metastatic pancreatic cancer. While being more effective, these regiments have a considerable degree of toxicity. Choosing the right treatment for any individual will be the next major challenge treating patients with pancreatic cancer.
Collapse
Affiliation(s)
- C Benedikt Westphalen
- a Department of Internal Medicine III and Comprehensive Cancer Center , Klinikum Grosshadern, Ludwig-Maximilians-University of Munich , Munich , Germany
| | - Stephan Kruger
- a Department of Internal Medicine III and Comprehensive Cancer Center , Klinikum Grosshadern, Ludwig-Maximilians-University of Munich , Munich , Germany
| | - Michael Haas
- a Department of Internal Medicine III and Comprehensive Cancer Center , Klinikum Grosshadern, Ludwig-Maximilians-University of Munich , Munich , Germany
| | - Volker Heinemann
- a Department of Internal Medicine III and Comprehensive Cancer Center , Klinikum Grosshadern, Ludwig-Maximilians-University of Munich , Munich , Germany
| | - Stefan Boeck
- a Department of Internal Medicine III and Comprehensive Cancer Center , Klinikum Grosshadern, Ludwig-Maximilians-University of Munich , Munich , Germany
| |
Collapse
|
760
|
Comment on: ‘Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort’. Br J Cancer 2016; 114:e8. [PMID: 27124336 PMCID: PMC4891507 DOI: 10.1038/bjc.2016.69] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
761
|
Ur Rehman SS, Lim K, Wang-Gillam A. Nanoliposomal irinotecan plus fluorouracil and folinic acid: a new treatment option in metastatic pancreatic cancer. Expert Rev Anticancer Ther 2016; 16:485-92. [PMID: 27043737 DOI: 10.1080/14737140.2016.1174581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease with half of patients diagnosed in the metastatic setting. Until recently, patients after progression on front-line gemcitabine-based regimen had no standard second-line option, although flouropyrimidine-based regimens were frequently used in this setting. Encapsulation of chemotherapeutics in liposomal formulation is an effective way of prolonging drug deposition thereby enhancing cytotoxic efficacy. In a large phase III randomized trial on metastatic PDAC patients who progressed after gemcitabine-based chemotherapy, a novel nanoliposome-encapsulated irinotecan (PEP02, MM-398, nal-IRI, Onivyde, Merrimack, Boston, US) plus fluorouracil and folinic acid demonstrated a significant survival advantage compared to fluorouracil and folinic acid alone. This pivotal study led to the recent FDA approval of nanoliposomal irinotecan in patients with metastatic PDAC. In this article, we will review the literature regarding existing treatment options for metastatic PDAC, focusing specifically on nanoliposomal irinotecan in the clinical setting and its future implication.
Collapse
Affiliation(s)
- Sana Saif Ur Rehman
- a Division of Oncology, Department of Medicine , Washington University in St. Louis , St. Louis , MO , USA
| | - Kian Lim
- a Division of Oncology, Department of Medicine , Washington University in St. Louis , St. Louis , MO , USA.,b Siteman Cancer Center , Washington University School of Medicine , St. Louis , MO , USA
| | - Andrea Wang-Gillam
- a Division of Oncology, Department of Medicine , Washington University in St. Louis , St. Louis , MO , USA.,b Siteman Cancer Center , Washington University School of Medicine , St. Louis , MO , USA
| |
Collapse
|
762
|
Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH, Neoptolemos JP. Pancreatic cancer. Nat Rev Dis Primers 2016; 2:16022. [PMID: 27158978 DOI: 10.1038/nrdp.2016.22] [Citation(s) in RCA: 1223] [Impact Index Per Article: 135.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer is a major cause of cancer-associated mortality, with a dismal overall prognosis that has remained virtually unchanged for many decades. Currently, prevention or early diagnosis at a curable stage is exceedingly difficult; patients rarely exhibit symptoms and tumours do not display sensitive and specific markers to aid detection. Pancreatic cancers also have few prevalent genetic mutations; the most commonly mutated genes are KRAS, CDKN2A (encoding p16), TP53 and SMAD4 - none of which are currently druggable. Indeed, therapeutic options are limited and progress in drug development is impeded because most pancreatic cancers are complex at the genomic, epigenetic and metabolic levels, with multiple activated pathways and crosstalk evident. Furthermore, the multilayered interplay between neoplastic and stromal cells in the tumour microenvironment challenges medical treatment. Fewer than 20% of patients have surgically resectable disease; however, neoadjuvant therapies might shift tumours towards resectability. Although newer drug combinations and multimodal regimens in this setting, as well as the adjuvant setting, appreciably extend survival, ∼80% of patients will relapse after surgery and ultimately die of their disease. Thus, consideration of quality of life and overall survival is important. In this Primer, we summarize the current understanding of the salient pathophysiological, molecular, translational and clinical aspects of this disease. In addition, we present an outline of potential future directions for pancreatic cancer research and patient management.
Collapse
Affiliation(s)
- Jorg Kleeff
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Duncan Building, Daulby Street, Liverpool L69 3GA, UK
- Department of General, Visceral and Pediatric Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Murray Korc
- Departments of Medicine, and Biochemistry and Molecular Biology, Indiana University School of Medicine, the Melvin and Bren Simon Cancer Center, and the Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Minoti Apte
- SWS Clinical School, University of New South Wales, and Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Colin D Johnson
- University Surgical Unit, University Hospital Southampton, Southampton, UK
| | - Andrew V Biankin
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, UK
| | - Rachel E Neale
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Margaret Tempero
- UCSF Pancreas Center, University of California San Francisco - Mission Bay Campus/Mission Hall, San Francisco, California, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, New York, USA
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John P Neoptolemos
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Duncan Building, Daulby Street, Liverpool L69 3GA, UK
| |
Collapse
|
763
|
|
764
|
|
765
|
Bupathi M, Ahn DH, Wu C, Ciombor KK, Stephens JA, Reardon J, Goldstein DA, Bekaii-Saab T. Modified irinotecan and infusional 5-fluorouracil (mFOLFIRI) in patients with refractory advanced pancreas cancer (APC): a single-institution experience. Med Oncol 2016; 33:37. [PMID: 26995224 PMCID: PMC4976592 DOI: 10.1007/s12032-016-0753-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death. Recently, MM-398 (nanoliposomal irinotecan) was shown to be associated with significant improvement in outcome measures with acceptable toxicities when combined with 5-fluorouracil (5-FU)/leucovorin (LV) compared to 5-FU/LV alone in patients failing one line of gemcitabine-based therapy. There is a paucity of data evaluating the role of irinotecan in combination with 5FU in advanced pancreas cancer (APC). We performed a retrospective analysis of all patients who received mFOLFIRI (minus bolus 5FU and LV). All patients with metastatic disease who had failed at least one line of gemcitabine-based therapy prior to receiving mFOLFIRI were included in this study. Descriptive statistics were used to assess the continuous variables and adverse events (AEs), and Kaplan-Meier methods were used to calculate the median progression-free survival (PFS) and overall survival (OS). Forty patients were included in this analysis. Patients received 1-5 lines of prior therapy (25 % with more than 3 lines of prior therapy). The mean age at diagnosis was 60, and 98 % had ECOG of 1. The mean CA 19-9 at the start of therapy was 33,169 U/ml. The median PFS was 2.59 months [95 % confidence interval (CI) (1.90, 3.54)], and OS was 4.75 months [95 % CI (3.14, 8.98)]. The most common AEs included fatigue (98 %), neuropathy (83 %), anorexia (68 %), nausea (60 %) and constipation (55 %). Grade 3 toxicities included fatigue (13 %) and rash (3 %). There were no observed grade 4 toxicities. In this single-institution retrospective analysis, mFOLFIRI was found to be both tolerable and relatively effective in a heavily pretreated patient population with APC. Future prospective studies should consider evaluating the role of mFOLFIRI in refractory APC.
Collapse
Affiliation(s)
- M Bupathi
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - D H Ahn
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - C Wu
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - K K Ciombor
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA
| | - J A Stephens
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - J Reardon
- Department of Pharmacy, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University, Columbus, OH, USA
| | - D A Goldstein
- Winship Cancer Institute, Emory University, 1365-C Clifton Rd NE, Atlanta, GA, USA
- Davidoff Cancer Center, Rabin Medical Center, Derech Ze'ev Jabotinsky 39, 4941492, Petach Tikva, Israel
| | - T Bekaii-Saab
- Department of Medical Oncology, Richard Solove Research Institute and James Cancer Hospital, The Ohio State University Wexner Medical Center, A454 Startling Loving Hall, 320 West 10th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
766
|
Indolfi L, Ligorio M, Ting DT, Xega K, Tzafriri AR, Bersani F, Aceto N, Thapar V, Fuchs BC, Deshpande V, Baker AB, Ferrone CR, Haber DA, Langer R, Clark JW, Edelman ER. A tunable delivery platform to provide local chemotherapy for pancreatic ductal adenocarcinoma. Biomaterials 2016; 93:71-82. [PMID: 27082874 DOI: 10.1016/j.biomaterials.2016.03.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/06/2016] [Accepted: 03/28/2016] [Indexed: 02/09/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most devastating and painful cancers. It is often highly resistant to therapy owing to inherent chemoresistance and the desmoplastic response that creates a barrier of fibrous tissue preventing transport of chemotherapeutics into the tumor. The growth of the tumor in pancreatic cancer often leads to invasion of other organs and partial or complete biliary obstruction, inducing intense pain for patients and necessitating tumor resection or repeated stenting. Here, we have developed a delivery device to provide enhanced palliative therapy for pancreatic cancer patients by providing high concentrations of chemotherapeutic compounds locally at the tumor site. This treatment could reduce the need for repeated procedures in advanced PDAC patients to debulk the tumor mass or stent the obstructed bile duct. To facilitate clinical translation, we created the device out of currently approved materials and drugs. We engineered an implantable poly(lactic-co-glycolic)-based biodegradable device that is able to linearly release high doses of chemotherapeutic drugs for up to 60 days. We created five patient-derived PDAC cell lines and tested their sensitivity to approved chemotherapeutic compounds. These in vitro experiments showed that paclitaxel was the most effective single agent across all cell lines. We compared the efficacy of systemic and local paclitaxel therapy on the patient-derived cell lines in an orthotopic xenograft model in mice (PDX). In this model, we found up to a 12-fold increase in suppression of tumor growth by local therapy in comparison to systemic administration and reduce retention into off-target organs. Herein, we highlight the efficacy of a local therapeutic approach to overcome PDAC chemoresistance and reduce the need for repeated interventions and biliary obstruction by preventing local tumor growth. Our results underscore the urgent need for an implantable drug-eluting platform to deliver cytotoxic agents directly within the tumor mass as a novel therapeutic strategy for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Laura Indolfi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Matteo Ligorio
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Health Sciences, University of Genoa, Genoa, Italy
| | - David T Ting
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kristina Xega
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Francesca Bersani
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicola Aceto
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vishal Thapar
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bryan C Fuchs
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vikram Deshpande
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Cristina R Ferrone
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A Haber
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Robert Langer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey W Clark
- Departments of Surgery, Medicine, and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
767
|
New drug for pancreatic cancer highlights the dual effect of regulatory approvals. Nat Rev Clin Oncol 2016; 13:205-6. [DOI: 10.1038/nrclinonc.2016.22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
768
|
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
|
769
|
Chen J, Diamond DJ, Manuel ER. Developing Effective Salmonella-based Approaches to Treat Pancreatic Cancer. ACTA ACUST UNITED AC 2016; 6:1-2. [PMID: 26925306 PMCID: PMC4764990 DOI: 10.4172/2165-7092.1000167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jeremy Chen
- Department of Experimental Therapeutics, City of Hope, Duarte CA, USA
| | - Don J Diamond
- Department of Experimental Therapeutics, City of Hope, Duarte CA, USA
| | - Edwin R Manuel
- Department of Experimental Therapeutics, City of Hope, Duarte CA, USA
| |
Collapse
|
770
|
Chemotherapy: NAPOLI-1: winning scoop for metastatic pancreatic cancer. Nat Rev Clin Oncol 2015; 13:2-3. [PMID: 26667974 DOI: 10.1038/nrclinonc.2015.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
771
|
Ueno M, Okusaka T, Omuro Y, Isayama H, Fukutomi A, Ikeda M, Mizuno N, Fukuzawa K, Furukawa M, Iguchi H, Sugimori K, Furuse J, Shimada K, Ioka T, Nakamori S, Baba H, Komatsu Y, Takeuchi M, Hyodo I, Boku N. A randomized phase II study of S-1 plus oral leucovorin versus S-1 monotherapy in patients with gemcitabine-refractory advanced pancreatic cancer. Ann Oncol 2015; 27:502-8. [PMID: 26681680 PMCID: PMC4769993 DOI: 10.1093/annonc/mdv603] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/01/2015] [Indexed: 12/27/2022] Open
Abstract
This is the first phase II trial in which adding oral leucovorin (LV) to S-1 (SL) significantly prolonged progression-free survival (PFS) when compared with S-1 monotherapy (S) in patients with gemcitabine-refractory advanced pancreatic cancer (PC). The significantly better PFS and disease control rate with SL than with S suggest that the antitumor activity of S-1 is enhanced by LV in advanced PC. Background We evaluated the efficacy and toxicity of adding oral leucovorin (LV) to S-1 when compared with S-1 monotherapy in patients with gemcitabine-refractory pancreatic cancer (PC). Patients and methods Gemcitabine-refractory PC patients were randomly assigned in a 1:1 ratio to receive S-1 at 40, 50, or 60 mg according to body surface area plus LV 25 mg, both given orally twice daily for 1 week, repeated every 2 weeks (SL group), or S-1 monotherapy at the same dose as the SL group for 4 weeks, repeated every 6 weeks (S-1 group). The primary end point was progression-free survival (PFS). Results Among 142 patients enrolled, 140 were eligible for efficacy assessment (SL: n = 69 and S-1: n = 71). PFS was significantly longer in the SL group than in the S-1 group [median PFS, 3.8 versus 2.7 months; hazard ratio (HR), 0.56; 95% confidence interval (CI), 0.37–0.85; P = 0.003]). The disease control rate was significantly higher in the SL group than in the S-1 group (91% versus 72%; P = 0.004). Overall survival (OS) was similar in both groups (median OS, 6.3 versus 6.1 months; HR, 0.82; 95% CI, 0.54–1.22; P = 0.463). After adjusting for patient background factors in a multivariate analysis, OS tended to be better in the SL group (HR, 0.71; 95% CI, 0.47–1.07; P = 0.099). Both treatments were well tolerated, although gastrointestinal toxicities were slightly more severe in the SL group. Conclusion The addition of LV to S-1 significantly improved PFS in patients with gemcitabine-refractory advanced PC, and a phase III trial has been initiated in a similar setting. Clinical trials number Japan Pharmaceutical Information Center: JapicCTI-111554.
Collapse
Affiliation(s)
- M Ueno
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Yokohama
| | - T Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo
| | - Y Omuro
- Department of Medical Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo
| | - H Isayama
- Department of Gastroenterology, The University of Tokyo, Graduate School of Medicine, Tokyo
| | - A Fukutomi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka
| | - M Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa
| | - N Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya
| | - K Fukuzawa
- Department of Surgery, Oita Red Cross Hospital, Oita
| | - M Furukawa
- Department of Gastroenterology, National Kyushu Cancer Center, Fukuoka
| | - H Iguchi
- Department of Gastroenterology, National Hospital Organization Shikoku Cancer Center, Matsuyama
| | - K Sugimori
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama
| | - J Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, Tokyo
| | - K Shimada
- Department of Medical Oncology, Saitama Medical University International Medical Center, Saitama
| | - T Ioka
- Department of Hepatobiliary and Pancreatic Oncology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka
| | - S Nakamori
- Hepato-biliary-pancreatic Surgery, National Hospital Organization Osaka National Hospital, Osaka
| | - H Baba
- Department of Gastroenterological Surgery, Kumamoto University, Kumamoto
| | - Y Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo
| | - M Takeuchi
- Department of Clinical Medicine (Biostatistics and Pharmaceutical Medicine), Kitasato University School of Pharmacy, Tokyo
| | - I Hyodo
- Division of Gastroenterology, University of Tsukuba, Tsukuba
| | - N Boku
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|