751
|
Facchinetti F, Tiseo M, Di Maio M, Graziano P, Bria E, Rossi G, Novello S. Tackling ALK in non-small cell lung cancer: the role of novel inhibitors. Transl Lung Cancer Res 2016; 5:301-21. [PMID: 27413712 DOI: 10.21037/tlcr.2016.06.10] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Crizotinib is an oral inhibitor of anaplastic lymphoma kinase (ALK) with remarkable clinical activity in patients suffering from ALK-rearranged non-small cell lung cancer (NSCLC), accounting to its superiority compared to chemotherapy. Unfortunately, virtually all ALK-rearranged tumors acquire resistance to crizotinib, frequently within one year since the treatment initiation. To date, therapeutic strategies to overcome crizotinib resistance have focused on the use of more potent and structurally different compounds. Second-generation ALK inhibitors such as ceritinib (LDK378), alectinib (CH5424802/RO5424802) and brigatinib (AP26113) have shown relevant clinical activity, consequently fostering their rapid clinical development and their approval by health agencies. The third-generation inhibitor lorlatinib (PF-06463922), selectively active against ALK and ROS1, harbors impressive biological potency; its efficacy in reversing resistance to crizotinib and to other ALK inhibitors is being proven by early clinical trials. The NTRK1-3 and ROS1 inhibitor entrectinib (RXDX-101) has been reported to act against NSCLC harboring ALK fusion proteins too. Despite the quick development of these novel agents, several issues remain to be discussed in the treatment of patients suffering from ALK-rearranged NSCLC. This position paper will discuss the development, the current evidence and approvals, as long as the future perspectives of new ALK inhibitors beyond crizotinib. Clinical behaviors of ALK-rearranged NSCLC vary significantly among patients and differential molecular events responsible of crizotinib resistance account for the most important quote of this heterogeneity. The precious availability of a wide range of active anti-ALK compounds should be approached in a critical and careful perspective, in order to develop treatment strategies tailored on the disease evolution of every single patient.
Collapse
Affiliation(s)
- Francesco Facchinetti
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Marcello Tiseo
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Massimo Di Maio
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Paolo Graziano
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Emilio Bria
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Giulio Rossi
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| | - Silvia Novello
- 1 INSERM U981, Gustave Roussy Cancer Campus, Université Paris-Sud, Villejuif, France ; 2 Medical Oncology Unit, University Hospital of Parma, Parma, Italy ; 3 Department of Oncology, AOU San Luigi (Orbassano), University of Turin, Italy ; 4 Medical Oncology, AO Ordine Mauriziano, Turin, Italy ; 5 Division of Pathology, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy ; 6 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy ; 7 Operative Unit of Pathology, Azienda USL Valle d'Aosta, Aosta, Italy
| |
Collapse
|
752
|
Zhang YC, Zhou Q, Wu YL. Efficacy of crizotinib in first-line treatment of adults with ALK-positive advanced NSCLC. Expert Opin Pharmacother 2016; 17:1693-701. [PMID: 27359268 DOI: 10.1080/14656566.2016.1208171] [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: 01/09/2023]
Abstract
INTRODUCTION The treatment of advanced non-small cell lung cancer (NSCLC) has evolved from palliative cytotoxic chemotherapy to precise medicine based on genetic alternations over the last decade. Anaplastic lymphoma kinase (ALK) rearrangement characterizes a molecular subset of NSCLC with an impressive response to crizotinib. AREAS COVERED To analyze the efficacy of crizotinib in first-line treatment of adults with advanced ALK-positive NSCLC, updated data on development and recent advances of first-line crizotinib in this subset population are reviewed. EXPERT OPINION To date, crizotinib should be established as a standard of care in previously untreated advanced NSCLC with ALK-rearrangement. However, the efficacy of first-line crizotinib is limited by acquired resistance. Second generation ALK inhibitors have demonstrated clinical activity in both crizotinib-refractory and crizotinib naïve setting. How to maximize first-line benefit for advanced ALK-positive NSCLC remains challenging. Combinational strategy, advances in companion diagnostics and optimization of ALK inhibitors might contribute to improve outcome in this subset of patients in future.
Collapse
Affiliation(s)
- Yi-Chen Zhang
- a Graduate School , Southern Medical University , Guangzhou , PR China.,b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| | - Qing Zhou
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| | - Yi-Long Wu
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| |
Collapse
|
753
|
Patel JN. Cancer pharmacogenomics, challenges in implementation, and patient-focused perspectives. Pharmgenomics Pers Med 2016; 9:65-77. [PMID: 27471406 PMCID: PMC4948716 DOI: 10.2147/pgpm.s62918] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer pharmacogenomics is an evolving landscape and has the potential to significantly impact cancer care and precision medicine. Harnessing and understanding the genetic code of both the patient (germline) and the tumor (somatic) provides the opportunity for personalized dose and therapy selection for cancer patients. While germline DNA is useful in understanding the pharmacokinetic and pharmacodynamic disposition of a drug, somatic DNA is particularly useful in identifying drug targets and predicting drug response. Molecular profiling of somatic DNA has resulted in the current breadth of targeted therapies available, expanding the armamentarium to battle cancer. This review provides an update on cancer pharmacogenomics and genomics-based medicine, challenges in applying pharmacogenomics to the clinical setting, and patient perspectives on the use of pharmacogenomics to personalize cancer therapy.
Collapse
Affiliation(s)
- Jai N Patel
- Department of Cancer Pharmacology, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| |
Collapse
|
754
|
Facchinetti F, Loriot Y, Kuo MS, Mahjoubi L, Lacroix L, Planchard D, Besse B, Farace F, Auger N, Remon J, Scoazec JY, André F, Soria JC, Friboulet L. Crizotinib-Resistant ROS1 Mutations Reveal a Predictive Kinase Inhibitor Sensitivity Model for ROS1- and ALK-Rearranged Lung Cancers. Clin Cancer Res 2016; 22:5983-5991. [DOI: 10.1158/1078-0432.ccr-16-0917] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 11/16/2022]
|
755
|
Lee DW, Lee KH, Kim JW, Keam B. Molecular Targeted Therapies for the Treatment of Leptomeningeal Carcinomatosis: Current Evidence and Future Directions. Int J Mol Sci 2016; 17:ijms17071074. [PMID: 27399673 PMCID: PMC4964450 DOI: 10.3390/ijms17071074] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 12/16/2022] Open
Abstract
Leptomeningeal carcinomatosis (LMC) is the multifocal seeding of cerebrospinal fluid and leptomeninges by malignant cells. The incidence of LMC is approximately 5% in patients with malignant tumors overall and the rate is increasing due to increasing survival time of cancer patients. Eradication of the disease is not yet possible, so the treatment goals of LMC are to improve neurologic symptoms and to prolong survival. A standard treatment for LMC has not been established due to low incidences of LMC, the rapidly progressing nature of the disease, heterogeneous populations with LMC, and a lack of randomized clinical trial results. Treatment options for LMC include intrathecal chemotherapy, systemic chemotherapy, and radiation therapy, but the prognoses remain poor with a median survival of <3 months. Recently, molecular targeted agents have been applied in the clinic and have shown groundbreaking results in specific patient groups epidermal growth factor receptor (EGFR)-targeted therapy or an anaplastic lymphoma kinase (ALK) inhibitor in lung cancer, human epidermal growth factor receptor 2 (HER2)-directed therapy in breast cancer, and CD20-targeted therapy in B cell lymphoma). Moreover, there are results indicating that the use of these agents under proper dose and administration routes can be effective for managing LMC. In this article, we review molecular targeted agents for managing LMC.
Collapse
Affiliation(s)
- Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Jin Wook Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea.
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
756
|
Yoshida T, Oya Y, Tanaka K, Shimizu J, Horio Y, Kuroda H, Sakao Y, Hida T, Yatabe Y. Clinical impact of crizotinib on central nervous system progression in ALK-positive non-small lung cancer. Lung Cancer 2016; 97:43-7. [DOI: 10.1016/j.lungcan.2016.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 12/31/2022]
|
757
|
Wicki A, Mandalà M, Massi D, Taverna D, Tang H, Hemmings BA, Xue G. Acquired Resistance to Clinical Cancer Therapy: A Twist in Physiological Signaling. Physiol Rev 2016; 96:805-29. [DOI: 10.1152/physrev.00024.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although modern therapeutic strategies have brought significant progress to cancer care in the last 30 years, drug resistance to targeted monotherapies has emerged as a major challenge. Aberrant regulation of multiple physiological signaling pathways indispensable for developmental and metabolic homeostasis, such as hyperactivation of pro-survival signaling axes, loss of suppressive regulations, and impaired functionalities of the immune system, have been extensively investigated aiming to understand the diversity of molecular mechanisms that underlie cancer development and progression. In this review, we intend to discuss the molecular mechanisms of how conventional physiological signal transduction confers to acquired drug resistance in cancer patients. We will particularly focus on protooncogenic receptor kinase inhibition-elicited tumor cell adaptation through two major core downstream signaling cascades, the PI3K/Akt and MAPK pathways. These pathways are crucial for cell growth and differentiation and are frequently hyperactivated during tumorigenesis. In addition, we also emphasize the emerging roles of the deregulated host immune system that may actively promote cancer progression and attenuate immunosurveillance in cancer therapies. Understanding these mechanisms may help to develop more effective therapeutic strategies that are able to keep the tumor in check and even possibly turn cancer into a chronic disease.
Collapse
Affiliation(s)
- Andreas Wicki
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Mario Mandalà
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Daniela Massi
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Daniela Taverna
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Huifang Tang
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Brian A. Hemmings
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Gongda Xue
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| |
Collapse
|
758
|
Yeh I. Recent advances in molecular genetics of melanoma progression: implications for diagnosis and treatment. F1000Res 2016; 5. [PMID: 27408703 PMCID: PMC4926755 DOI: 10.12688/f1000research.8247.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2016] [Indexed: 11/20/2022] Open
Abstract
According to the multi-step carcinogenesis model of cancer, initiation results in a benign tumor and subsequent genetic alterations lead to tumor progression and the acquisition of the hallmarks of cancer. This article will review recent discoveries in our understanding of initiation and progression in melanocytic neoplasia and the impact on diagnostic dermatopathology.
Collapse
Affiliation(s)
- Iwei Yeh
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
759
|
Campo M, Al-Halabi H, Khandekar M, Shaw AT, Sequist LV, Willers H. Integration of Stereotactic Body Radiation Therapy With Tyrosine Kinase Inhibitors in Stage IV Oncogene-Driven Lung Cancer. Oncologist 2016; 21:964-73. [PMID: 27354669 DOI: 10.1634/theoncologist.2015-0508] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/18/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED : Genotype-based selection of patients for targeted therapies has had a substantial impact on the treatment of non-small cell lung cancers (NSCLCs). Tyrosine kinase inhibitors (TKIs) directed at cancers driven by oncogenes, such as epidermal growth factor receptor mutations or anaplastic lymphoma kinase rearrangements, often achieve dramatic responses and result in prolonged survival compared with chemotherapy. However, TKI resistance invariably develops. Disease progression can be limited to only one or a few sites and might not be symptomatic, raising the important question of whether this type of oligoprogression warrants a change in systemic therapy or consideration of local treatment. Recent clinical observations suggest a growing role for stereotactic body radiation therapy (SBRT) in the treatment of oligoprogressive and perhaps even oligopersistent disease (primary and/or metastases) in oncogene-driven NSCLC. SBRT might allow patients to continue with existing TKI treatments longer and delay the need to switch to other systemic options. We review the current data with regard to the use of SBRT for metastatic NSCLC and particularly oncogene-driven disease. Although there is great promise in the marriage of targeted therapies with SBRT, prospective data are urgently needed. In the meantime, such strategies are being used in carefully selected patients, with risk-adapted SBRT dose-fractionation regimens used to optimize the therapeutic index. IMPLICATIONS FOR PRACTICE Stereotactic body radiation therapy (SBRT) or SBRT-like treatments are increasingly being used for oligoprogression in patients with oncogene-driven non-small cell lung cancer. This approach allows patients to extend the duration of tyrosine kinase inhibitor therapy and has the potential to prolong survival times. Careful patient selection and risk-adapted radiation dosing is of critical importance to minimize toxicity and preserve patient quality of life.
Collapse
Affiliation(s)
- Meghan Campo
- Hematology/Oncology Fellowship Program, Dana-Farber/Partners CancerCare, Boston, Massachusetts, USA
| | - Hani Al-Halabi
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Melin Khandekar
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alice T Shaw
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lecia V Sequist
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
760
|
Cortinovis D, Abbate M, Bidoli P, Capici S, Canova S. Targeted therapies and immunotherapy in non-small-cell lung cancer. Ecancermedicalscience 2016; 10:648. [PMID: 27433281 PMCID: PMC4929979 DOI: 10.3332/ecancer.2016.648] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Indexed: 01/23/2023] Open
Abstract
Non-small-cell lung cancer is still considered a difficult disease to manage because of its aggressiveness and resistance to common therapies. Chemotherapy remains the gold standard in nearly 80% of lung cancers, but clinical outcomes are discouraging, and the impact on median overall survival (OS) barely reaches 12 months. At the end of the last century, the discovery of oncogene-driven tumours completely changed the therapeutic landscape in lung cancers, harbouring specific gene mutations/translocations. Epidermal growth factors receptor (EGFR) common mutations first and anaplastic lymphoma kinase (ALK) translocations later led new insights in lung cancer biology knowledge. The use of specific tyrosine kinases inhibitors overturned the biological behaviour of EGFR mutation positive tumours and became a preclinical model to understand the heterogeneity of lung cancers and the mechanisms of drug resistance. In this review, we summarise the employment of targeted agents against the most representative biomolecular alterations and provide some criticisms of the therapeutic strategies.
Collapse
Affiliation(s)
- D Cortinovis
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - M Abbate
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - P Bidoli
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - S Capici
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| | - S Canova
- Medical Oncology Unit, AOU San Gerardo, via Giambattista Pergolesi 33, 20900 Monza, Italy
| |
Collapse
|
761
|
Zaarour M, Nazha B, Weerasinghe C, Moussaly E, Terjanian T. Anaplastic lymphoma kinase inhibitors in elderly patients with advanced non-small cell lung cancer. Expert Rev Anticancer Ther 2016; 16:877-83. [PMID: 27328177 DOI: 10.1080/14737140.2016.1204235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths in the USA and worldwide. At diagnosis, half of the patients are over 70 years of age, and most present with advanced disease for which chemotherapy provides modest benefit with significant toxicity. Older patients often have more comorbidities than their younger counterparts and tend to be excluded from clinical trials. AREAS COVERED A small percentage (less than 7%) of patients with NSCLC have the anaplastic lymphoma kinase (ALK) rearrangement. Compared to the general NSCLC population, this clinically distinct group has a relatively younger median age of 51 years at diagnosis. As such, elderly patients with ALK-positive disease are both a minority within this group and are expected to be underrepresented in clinical trials. Expert commentary: Despite promising results in the general population, data about the efficacy and safety of ALK inhibitors in the elderly population remains scarce. In our review, we briefly discuss the current evidence of ALK inhibitors in the general population and we shed light on this subgroup of elderly patients with advanced ALK-positive disease.
Collapse
Affiliation(s)
- Mazen Zaarour
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Bassel Nazha
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Chanudi Weerasinghe
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Elias Moussaly
- a Department of Medicine , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| | - Terenig Terjanian
- b Department of Medicine, Division of Hematology/Oncology , Staten Island University Hospital Northwell Health , Staten Island , NY , USA
| |
Collapse
|
762
|
Hong AL, Tseng YY, Cowley GS, Jonas O, Cheah JH, Kynnap BD, Doshi MB, Oh C, Meyer SC, Church AJ, Gill S, Bielski CM, Keskula P, Imamovic A, Howell S, Kryukov GV, Clemons PA, Tsherniak A, Vazquez F, Crompton BD, Shamji AF, Rodriguez-Galindo C, Janeway KA, Roberts CWM, Stegmaier K, van Hummelen P, Cima MJ, Langer RS, Garraway LA, Schreiber SL, Root DE, Hahn WC, Boehm JS. Integrated genetic and pharmacologic interrogation of rare cancers. Nat Commun 2016; 7:11987. [PMID: 27329820 PMCID: PMC4917959 DOI: 10.1038/ncomms11987] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023] Open
Abstract
Identifying therapeutic targets in rare cancers remains challenging due to the paucity of established models to perform preclinical studies. As a proof-of-concept, we developed a patient-derived cancer cell line, CLF-PED-015-T, from a paediatric patient with a rare undifferentiated sarcoma. Here, we confirm that this cell line recapitulates the histology and harbours the majority of the somatic genetic alterations found in a metastatic lesion isolated at first relapse. We then perform pooled CRISPR-Cas9 and RNAi loss-of-function screens and a small-molecule screen focused on druggable cancer targets. Integrating these three complementary and orthogonal methods, we identify CDK4 and XPO1 as potential therapeutic targets in this cancer, which has no known alterations in these genes. These observations establish an approach that integrates new patient-derived models, functional genomics and chemical screens to facilitate the discovery of targets in rare cancers.
Collapse
Affiliation(s)
- Andrew L. Hong
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Yuen-Yi Tseng
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Glenn S. Cowley
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Oliver Jonas
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Jaime H. Cheah
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Bryan D. Kynnap
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Mihir B. Doshi
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Coyin Oh
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Stephanie C. Meyer
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Alanna J. Church
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Shubhroz Gill
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Craig M. Bielski
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paula Keskula
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Alma Imamovic
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Sara Howell
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Gregory V. Kryukov
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Paul A. Clemons
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Aviad Tsherniak
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Francisca Vazquez
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Brian D. Crompton
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Alykhan F. Shamji
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Carlos Rodriguez-Galindo
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Katherine A. Janeway
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Charles W. M. Roberts
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Kimberly Stegmaier
- Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Paul van Hummelen
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Michael J. Cima
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Robert S. Langer
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, Massachusetts 02139, USA
| | - Levi A. Garraway
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Stuart L. Schreiber
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - David E. Root
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - William C. Hahn
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115, USA
| | - Jesse S. Boehm
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
763
|
Abstract
INTRODUCTION Anaplastic lymphoma kinase (ALK) rearrangements are present in about 5% of advanced non-small-cell lung cancer (NSCLC) patients. Despite the initial response, after a median of 1-2 years, ALK-positive patients developed an acquired resistance to the ALK-inhibitor crizotinib. Among the most promising second-generation ALK-inhibitors, alectinib is being investigated in crizotinib-naïve and -resistant ALK-positive NSCLC patients. AREAS COVERED The current state-of-the-art of ALK-inhibitors treatment, and in particular the role of alectinib in this setting, is reviewed and discussed. A structured search of bibliographic databases for peer-reviewed research literature and of main meetings using a focused review question was undertaken. Expert commentary: Alectinib reports promising results with a good safety profile, becoming a potentially very important option for ALK-translocated NSCLC patients. The preliminary results from the J-ALEX phase III randomized trial performed in ALK-rearranged NSCLC Japanese patients showed a better activity and tolerability of alectinib versus crizotinib.
Collapse
Affiliation(s)
- Antonio Rossi
- a Division of Medical Oncology , "S.G. Moscati" Hospital , Avellino , Italy
| |
Collapse
|
764
|
Schmidt KT, Chau CH, Price DK, Figg WD. Precision Oncology Medicine: The Clinical Relevance of Patient-Specific Biomarkers Used to Optimize Cancer Treatment. J Clin Pharmacol 2016; 56:1484-1499. [PMID: 27197880 DOI: 10.1002/jcph.765] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022]
Abstract
Precision medicine in oncology is the result of an increasing awareness of patient-specific clinical features coupled with the development of genomic-based diagnostics and targeted therapeutics. Companion diagnostics designed for specific drug-target pairs were the first to widely utilize clinically applicable tumor biomarkers (eg, HER2, EGFR), directing treatment for patients whose tumors exhibit a mutation susceptible to an FDA-approved targeted therapy (eg, trastuzumab, erlotinib). Clinically relevant germline mutations in drug-metabolizing enzymes and transporters (eg, TPMT, DPYD) have been shown to impact drug response, providing a rationale for individualized dosing to optimize treatment. The use of multigene expression-based assays to analyze an array of prognostic biomarkers has been shown to help direct treatment decisions, especially in breast cancer (eg, Oncotype DX). More recently, the use of next-generation sequencing to detect many potential "actionable" cancer molecular alterations is further shifting the 1 gene-1 drug paradigm toward a more comprehensive, multigene approach. Currently, many clinical trials (eg, NCI-MATCH, NCI-MPACT) are assessing novel diagnostic tools with a combination of different targeted therapeutics while also examining tumor biomarkers that were previously unexplored in a variety of cancer histologies. Results from ongoing trials such as the NCI-MATCH will help determine the clinical utility and future development of the precision-medicine approach.
Collapse
Affiliation(s)
- Keith T Schmidt
- Clinical Pharmacology Program, Office of the Clinical Director, NIH, Bethesda, MD, USA
| | - Cindy H Chau
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Douglas K Price
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - William D Figg
- Clinical Pharmacology Program, Office of the Clinical Director, NIH, Bethesda, MD, USA
- Molecular Pharmacology Section, Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
765
|
Giroux-Leprieur E, Fallet V, Cadranel J, Wislez M. Spotlight on crizotinib in the first-line treatment of ALK-positive advanced non-small-cell lung cancer: patients selection and perspectives. LUNG CANCER-TARGETS AND THERAPY 2016; 7:83-90. [PMID: 28210164 PMCID: PMC5310701 DOI: 10.2147/lctt.s99303] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Around 4% of advanced non-small-cell lung cancers (NSCLCs) have an ALK rearrangement at the time of diagnosis. This molecular feature is more frequent in young patients, with no/light smoking habit and with adenocarcinoma pathological subtype. Crizotinib is a tyrosine kinase inhibitor, targeting ALK, ROS1, RON, and MET. The preclinical efficacy results led to a fast-track clinical development. The US Food and Drug Administration (FDA) approval was achieved after the Phase I clinical trial in 2011 in ALK-rearranged advanced NSCLC progressing after a first-line treatment. In 2013, the randomized Phase III trial PROFILE-1007 confirmed the efficacy of crizotinib in ALK-rearranged NSCLC, compared to cytotoxic chemotherapy, in second-line setting or more. In 2014, the PROFILE-1014 trial showed the superiority of crizotinib in the first-line setting compared to the pemetrexed platinum doublet chemotherapy. The response rate was 74%, and the progression-free survival was 10.9 months with crizotinib. Based on these results, crizotinib received approval from the FDA and European Medicines Agency for first-line treatment of ALK-rearranged NSCLC. The various molecular mechanisms at the time of the progression (ALK mutations or amplification, ALK-independent mechanisms) encourage performing re-biopsy at the time of progression under crizotinib. The best treatment strategy at the progression (crizotinib continuation beyond progression, switch to second-generation tyrosine kinase inhibitors, or cytotoxic chemotherapy) depends on the phenotype of the progression, the molecular status, and the physical condition of the patient.
Collapse
Affiliation(s)
- Etienne Giroux-Leprieur
- Respiratory Diseases and Thoracic Oncology Department, APHP-Ambroise Paré Hospital, Boulogne-Billancourt, France; EA4340 Laboratory, UVSQ, Paris-Saclay University, France
| | - Vincent Fallet
- Respiratory Diseases Department, APHP - Tenon Hospital, Paris, France; Sorbonne University, GRC 04, UPMC Univ Paris 06, France
| | - Jacques Cadranel
- Respiratory Diseases Department, APHP - Tenon Hospital, Paris, France; Sorbonne University, GRC 04, UPMC Univ Paris 06, France
| | - Marie Wislez
- Respiratory Diseases Department, APHP - Tenon Hospital, Paris, France; Sorbonne University, GRC 04, UPMC Univ Paris 06, France
| |
Collapse
|
766
|
Abstract
Non-small cell lung carcinoma is a leading cause of cancer death worldwide. Understanding the molecular biology of survival and proliferation of cancer cells led to a new molecular classification of lung cancer and the development of targeted therapies with promising results. With the advances of image-guided biopsy techniques, tumor samples are becoming smaller, and the molecular testing techniques have to overcome the challenge of integrating the characterization of a panel of abnormalities including gene mutations, copy-number changes, and fusions in a reduced number of assays using only a small amount of genetic material. This article reviews the current knowledge about the most frequent actionable molecular abnormalities in non-small cell lung carcinoma, the new approaches of molecular analysis, and the implications of these findings in the context of clinical practice.
Collapse
|
767
|
Liu C, Ding L, Sun B, Wu S. Bilateral breast adenocarcinomas with EML4-ALK fusion in a patient with multiple metastases successfully treated with crizotinib: is lung the primary site? Onco Targets Ther 2016; 9:3589-3593. [PMID: 27366096 PMCID: PMC4913993 DOI: 10.2147/ott.s104583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Breast metastases from non-mammary cancers are rare, especially when they appear synchronously. Clinically, it is vitally important to accurately diagnose these patients, as this will directly influence their treatment and survival. We present a very rare and complex case of bilateral breast adenocarcinomas with an EML4-ALK fusion, which was diagnosed as bilateral breast metastases of non-small-cell lung cancer by immunohistochemistry and comprehensive genomic investigation. The patient was successfully treated with an ALK inhibitor (crizotinib); symptoms improved quickly after initiation of crizotinib therapy, and a partial response was observed after 3 months. The experience of diagnosis and treatment of this case indicates the importance and necessity of genomic investigations in such patients, and suggests that we need to consider the rare possibility of this kind of metastasis in order to provide optimal treatment.
Collapse
Affiliation(s)
- Chao Liu
- Department of Radiation Oncology, 307 Hospital of People’s Liberation Army, 307 Clinical College, Anhui Medical University, Beijing, People’s Republic of China
- Department of Radiation Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Lijuan Ding
- Department of Radiation Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Bing Sun
- Department of Radiation Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Shikai Wu
- Department of Radiation Oncology, 307 Hospital of People’s Liberation Army, 307 Clinical College, Anhui Medical University, Beijing, People’s Republic of China
- Department of Radiation Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
768
|
Marchetti A, Pace MV, Di Lorito A, Canarecci S, Felicioni L, D'Antuono T, Liberatore M, Filice G, Guetti L, Mucilli F, Buttitta F. Validation of a new algorithm for a quick and easy RT-PCR-based ALK test in a large series of lung adenocarcinomas: Comparison with FISH, immunohistochemistry and next generation sequencing assays. Lung Cancer 2016; 99:11-6. [PMID: 27565907 DOI: 10.1016/j.lungcan.2016.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Anaplastic Lymphoma Kinase (ALK) gene rearrangements have been described in 3-5% of lung adenocarcinomas (ADC) and their identification is essential to select patients for treatment with ALK tyrosine kinase inhibitors. For several years, fluorescent in situ hybridization (FISH) has been considered as the only validated diagnostic assay. Currently, alternative methods are commercially available as diagnostic tests. MATERIAL AND METHODS A series of 217 ADC comprising 196 consecutive resected tumors and 21 ALK FISH-positive cases from an independent series of 702 ADC were investigated. All specimens were screened by IHC (ALK-D5F3-CDx-Ventana), FISH (Vysis ALK Break-Apart-Abbott) and RT-PCR (ALK RGQ RT-PCR-Qiagen). Results were compared and discordant cases subjected to Next Generation Sequencing. RESULTS Thirty-nine of 217 samples were positive by the ALK RGQ RT-PCR assay, using a threshold cycle (Ct) cut-off ≤35.9, as recommended. Of these positive samples, 14 were negative by IHC and 12 by FISH. ALK RGQ RT-PCR/FISH discordant cases were analyzed by the NGS assay with results concordant with FISH data. In order to obtain the maximum level of agreement between FISH and ALK RGQ RT-PCR data, we introduced a new scoring algorithm based on the ΔCt value. A ΔCt cut-off level ≤3.5 was used in a pilot series. Then the algorithm was tested on a completely independent validation series. By using the new scoring algorithm and FISH as reference standard, the sensitivity and the specificity of the ALK RGQ RT-PCR(ΔCt) assay were 100% and 100%, respectively. CONCLUSIONS Our results suggest that the ALK RGQ RT-PCR test could be useful in clinical practice as a complementary assay in multi-test diagnostic algorithms or even, if our data will be confirmed in independent studies, as a standalone or screening test for the selection of patients to be treated with ALK inhibitors.
Collapse
Affiliation(s)
- Antonio Marchetti
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Maria Vittoria Pace
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Alessia Di Lorito
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Sara Canarecci
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Lara Felicioni
- Oncological and Cardiovascular Molecular Medicine Unit, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti, Chieti, Italy
| | - Tommaso D'Antuono
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Marcella Liberatore
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Giampaolo Filice
- Center of Predictive Molecular Medicine, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti-Pescara, Chieti, Italy
| | - Luigi Guetti
- Department of Surgery, University of Chieti, Chieti, Italy
| | - Felice Mucilli
- Department of Surgery, University of Chieti, Chieti, Italy
| | - Fiamma Buttitta
- Oncological and Cardiovascular Molecular Medicine Unit, Center for Excellence on Ageing and Translational Medicine (CeSI-MeT), University of Chieti, Chieti, Italy.
| |
Collapse
|
769
|
Syn NLX, Yong WP, Goh BC, Lee SC. Evolving landscape of tumor molecular profiling for personalized cancer therapy: a comprehensive review. Expert Opin Drug Metab Toxicol 2016; 12:911-22. [PMID: 27249175 DOI: 10.1080/17425255.2016.1196187] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Tumour molecular profiling has been at the crossroads of large-scale integrative genomic studies and major clinical trials over the past 5 years and has provided roadmaps for better disease stratification and therapeutic management. AREAS COVERED We review the landscape of precision oncology trials in Asia, Europe and the United States, and emerging insights gained from recently reported studies such as the SHIVA and CUSTOM trials. Changes in the molecular portraits of human cancers and the immune contexture of the tumor microenvironment during treatment may predict the course of tumor progression, including the development of treatment resistance. 'Liquid biopsy' approaches that harness circulating tumor cells, cell-free DNA and exosomes may provide a non-invasive means of monitoring the parent tumor in real-time. Several molecular signatures are being evaluated as biomarkers for emerging immunologic approaches, such as the mismatch-repair deficiency status and nonsynonymous mutation burden in anti-PD-1 immune checkpoint blockade. Finally, we review the current actionability and future clinical impact of multigene panel and next-generation sequencing (NGS)-based profiling. EXPERT OPINION In the future, molecular profiling may help to fulfill unmet needs for predictive biomarkers in novel immunotherapeutic approaches, while ongoing precision trials are laying the foundations for clinical uptake of NGS testing.
Collapse
Affiliation(s)
- Nicholas Li-Xun Syn
- a Department of Haematology-Oncology , National University Cancer Institute, National University Health System , Singapore , Singapore
| | - Wei-Peng Yong
- a Department of Haematology-Oncology , National University Cancer Institute, National University Health System , Singapore , Singapore
| | - Boon-Cher Goh
- a Department of Haematology-Oncology , National University Cancer Institute, National University Health System , Singapore , Singapore
| | - Soo-Chin Lee
- a Department of Haematology-Oncology , National University Cancer Institute, National University Health System , Singapore , Singapore
| |
Collapse
|
770
|
Profiling of Oncogenic Driver Events in Lung Adenocarcinoma Revealed MET Mutation as Independent Prognostic Factor. J Thorac Oncol 2016; 10:1292-1300. [PMID: 26098749 DOI: 10.1097/jto.0000000000000620] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Oncogenic driver mutations activating receptor tyrosine kinase pathways are promising predictive markers for targeted treatment. We investigated the mutation profile of an updated driver events list on receptor tyrosine kinase/RAS/PI3K axis and the clinicopathologic implications in a cohort of never-smoker predominated Chinese lung adenocarcinoma. METHODS We tested 154 lung adenocarcinomas and adenosquamous carcinomas for EGFR, KRAS, HER2, BRAF, PIK3CA, MET, NRAS, MAP2K1, and RIT1 mutations by polymerase chain reaction-direct sequencing. MET amplification and ALK and ROS1 translocations were assessed by fluorescent in situ hybridizations. MET and thyroid transcription factor-1 protein expressions were investigated by immunohistochemistry. RESULTS Seventy percent of lung adenocarcinomas carried actionable driver events. Alterations on EGFR (43%), KRAS (11.4%), ALK (6%), and MET (5.4%) were frequently found. ROS1 translocation and mutations involving BRAF, HER2, NRAS, and PIK3CA were also detected. No mutation was observed in RIT1 and MAP2K1. Patients with EGFR mutations had a favorable prognosis, whereas those with MET mutations had poorer overall survival. Multivariate analysis further demonstrated that MET mutation was an independent prognostic factor. Although MET protein expression was detected in 65% of lung adenocarcinoma, only 10% of the MET-immunohistochemistry positive tumors harbor MET DNA alterations that drove protein overexpression. Appropriate predictive biomarker is essential for selecting patients who might benefit from specific targeted therapy. CONCLUSION Actionable driver events can be detected in two thirds of lung adenocarcinoma. MET DNA alterations define a subset of patients with aggressive diseases that might potentially benefit from anti-MET targeted therapy. High negative predictive values of thyroid transcription factor-1 and MET expression suggest potential roles as surrogate markers for EGFR and/or MET mutations.
Collapse
|
771
|
Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, Haringsma HJ, Kurtz DM, Stehr H, Scherer F, Karlovich CA, Harding TC, Durkin KA, Otterson GA, Purcell WT, Camidge DR, Goldman JW, Sequist LV, Piotrowska Z, Wakelee HA, Neal JW, Alizadeh AA, Diehn M. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun 2016; 7:11815. [PMID: 27283993 PMCID: PMC4906406 DOI: 10.1038/ncomms11815] [Citation(s) in RCA: 488] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/03/2016] [Indexed: 12/20/2022] Open
Abstract
Circulating tumour DNA (ctDNA) analysis facilitates studies of tumour heterogeneity. Here we employ CAPP-Seq ctDNA analysis to study resistance mechanisms in 43 non-small cell lung cancer (NSCLC) patients treated with the third-generation epidermal growth factor receptor (EGFR) inhibitor rociletinib. We observe multiple resistance mechanisms in 46% of patients after treatment with first-line inhibitors, indicating frequent intra-patient heterogeneity. Rociletinib resistance recurrently involves MET, EGFR, PIK3CA, ERRB2, KRAS and RB1. We describe a novel EGFR L798I mutation and find that EGFR C797S, which arises in ∼33% of patients after osimertinib treatment, occurs in <3% after rociletinib. Increased MET copy number is the most frequent rociletinib resistance mechanism in this cohort and patients with multiple pre-existing mechanisms (T790M and MET) experience inferior responses. Similarly, rociletinib-resistant xenografts develop MET amplification that can be overcome with the MET inhibitor crizotinib. These results underscore the importance of tumour heterogeneity in NSCLC and the utility of ctDNA-based resistance mechanism assessment.
Collapse
Affiliation(s)
- Jacob J. Chabon
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | | | - Alexander F. Lovejoy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | - Mohammad S. Esfahani
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | - Aaron M. Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | | | - David M. Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA
| | - Henning Stehr
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
| | - Florian Scherer
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | | | | | - Kathleen A. Durkin
- Molecular Graphics and Computation Facility, College of Chemistry, University of California, Berkeley, California 94720, USA
| | | | - W. Thomas Purcell
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - D. Ross Camidge
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Jonathan W. Goldman
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Lecia V. Sequist
- Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zofia Piotrowska
- Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Heather A. Wakelee
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Joel W. Neal
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Ash A. Alizadeh
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, USA
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Maximilian Diehn
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Stanford Cancer Institute, Stanford University, Stanford, California 94305, USA
- Department of Radiation Oncology, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
772
|
Melosky B, Agulnik J, Albadine R, Banerji S, Bebb DG, Bethune D, Blais N, Butts C, Cheema P, Cheung P, Cohen V, Deschenes J, Ionescu DN, Juergens R, Kamel-Reid S, Laurie SA, Liu G, Morzycki W, Tsao MS, Xu Z, Hirsh V. Canadian consensus: inhibition of ALK-positive tumours in advanced non-small-cell lung cancer. ACTA ACUST UNITED AC 2016; 23:196-200. [PMID: 27330348 DOI: 10.3747/co.23.3120] [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: 12/22/2022]
Abstract
Anaplastic lymphoma kinase (alk) is an oncogenic driver in non-small-cell lung cancer (nsclc). Chromosomal rearrangements involving the ALK gene occur in up to 4% of nonsquamous nsclc patients and lead to constitutive activation of the alk signalling pathway. ALK-positive nsclc is found in relatively young patients, with a median age of 50 years. Patients frequently have brain metastasis. Targeted inhibition of the alk pathway prolongs progression-free survival in patients with ALK-positive advanced nsclc. The results of several recent clinical trials confirm the efficacy and safety benefit of crizotinib and ceritinib in this population. Canadian oncologists support the following consensus statement: All patients with advanced nonsquamous nsclc (excluding pure neuroendocrine carcinoma) should be tested for the presence of an ALK rearrangement. If an ALK rearrangement is present, treatment with a targeted alk inhibitor in the first-line setting is recommended. As patients become resistant to first-generation alk inhibitors, other treatments, including second-generation alk inhibitors can be considered.
Collapse
Affiliation(s)
- B Melosky
- British Columbia: BC Cancer Agency, Vancouver Centre, Vancouver, BC (Melosky, Ionescu)
| | - J Agulnik
- Quebec: Jewish General Hospital, McGill University, Montreal, QC (Agulnik); chum -Hôpital St-Luc, Montreal, QC (Albadine); chum -Hôpital Notre-Dame, Montreal, QC (Blais); Royal Victoria Hospital, Montreal, QC (Hirsh); Segal Cancer Centre and Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC (Cohen)
| | - R Albadine
- Quebec: Jewish General Hospital, McGill University, Montreal, QC (Agulnik); chum -Hôpital St-Luc, Montreal, QC (Albadine); chum -Hôpital Notre-Dame, Montreal, QC (Blais); Royal Victoria Hospital, Montreal, QC (Hirsh); Segal Cancer Centre and Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC (Cohen)
| | - S Banerji
- Manitoba: CancerCare Manitoba and University of Manitoba, Winnipeg, MB (Banerji)
| | - D G Bebb
- Alberta: Tom Baker Cancer Centre, Calgary, AB (Bebb); Cross Cancer Institute and University of Alberta, Edmonton, AB (Butts, Deschenes)
| | - D Bethune
- Nova Scotia: QEII Health Sciences Centre, Halifax, NS (Bethune, Morzycki, Xu)
| | - N Blais
- Quebec: Jewish General Hospital, McGill University, Montreal, QC (Agulnik); chum -Hôpital St-Luc, Montreal, QC (Albadine); chum -Hôpital Notre-Dame, Montreal, QC (Blais); Royal Victoria Hospital, Montreal, QC (Hirsh); Segal Cancer Centre and Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC (Cohen)
| | - C Butts
- Alberta: Tom Baker Cancer Centre, Calgary, AB (Bebb); Cross Cancer Institute and University of Alberta, Edmonton, AB (Butts, Deschenes)
| | - P Cheema
- Ontario: Sunnybrook Odette Cancer Centre, Toronto, ON (Cheema, Cheung); Juravinski Cancer Centre, Hamilton, ON (Juergens); University Health Network, Princess Margaret Cancer Centre, Toronto, ON (Kamel-Reid, Liu, Tsao); The Ottawa Hospital Cancer Centre, Ottawa, ON (Laurie)
| | - P Cheung
- Ontario: Sunnybrook Odette Cancer Centre, Toronto, ON (Cheema, Cheung); Juravinski Cancer Centre, Hamilton, ON (Juergens); University Health Network, Princess Margaret Cancer Centre, Toronto, ON (Kamel-Reid, Liu, Tsao); The Ottawa Hospital Cancer Centre, Ottawa, ON (Laurie)
| | - V Cohen
- Quebec: Jewish General Hospital, McGill University, Montreal, QC (Agulnik); chum -Hôpital St-Luc, Montreal, QC (Albadine); chum -Hôpital Notre-Dame, Montreal, QC (Blais); Royal Victoria Hospital, Montreal, QC (Hirsh); Segal Cancer Centre and Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC (Cohen)
| | - J Deschenes
- Alberta: Tom Baker Cancer Centre, Calgary, AB (Bebb); Cross Cancer Institute and University of Alberta, Edmonton, AB (Butts, Deschenes)
| | - D N Ionescu
- British Columbia: BC Cancer Agency, Vancouver Centre, Vancouver, BC (Melosky, Ionescu)
| | - R Juergens
- Ontario: Sunnybrook Odette Cancer Centre, Toronto, ON (Cheema, Cheung); Juravinski Cancer Centre, Hamilton, ON (Juergens); University Health Network, Princess Margaret Cancer Centre, Toronto, ON (Kamel-Reid, Liu, Tsao); The Ottawa Hospital Cancer Centre, Ottawa, ON (Laurie)
| | - S Kamel-Reid
- Ontario: Sunnybrook Odette Cancer Centre, Toronto, ON (Cheema, Cheung); Juravinski Cancer Centre, Hamilton, ON (Juergens); University Health Network, Princess Margaret Cancer Centre, Toronto, ON (Kamel-Reid, Liu, Tsao); The Ottawa Hospital Cancer Centre, Ottawa, ON (Laurie)
| | - S A Laurie
- British Columbia: BC Cancer Agency, Vancouver Centre, Vancouver, BC (Melosky, Ionescu)
| | - G Liu
- British Columbia: BC Cancer Agency, Vancouver Centre, Vancouver, BC (Melosky, Ionescu)
| | - W Morzycki
- Nova Scotia: QEII Health Sciences Centre, Halifax, NS (Bethune, Morzycki, Xu)
| | - M S Tsao
- Ontario: Sunnybrook Odette Cancer Centre, Toronto, ON (Cheema, Cheung); Juravinski Cancer Centre, Hamilton, ON (Juergens); University Health Network, Princess Margaret Cancer Centre, Toronto, ON (Kamel-Reid, Liu, Tsao); The Ottawa Hospital Cancer Centre, Ottawa, ON (Laurie)
| | - Z Xu
- Nova Scotia: QEII Health Sciences Centre, Halifax, NS (Bethune, Morzycki, Xu)
| | - V Hirsh
- Quebec: Jewish General Hospital, McGill University, Montreal, QC (Agulnik); chum -Hôpital St-Luc, Montreal, QC (Albadine); chum -Hôpital Notre-Dame, Montreal, QC (Blais); Royal Victoria Hospital, Montreal, QC (Hirsh); Segal Cancer Centre and Sir Mortimer B. Davis Jewish General Hospital, Montreal, QC (Cohen)
| |
Collapse
|
773
|
[Molecular epidemiology of the lung cancer]. Rev Mal Respir 2016; 33:675-681. [PMID: 27266901 DOI: 10.1016/j.rmr.2016.01.007] [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: 10/09/2015] [Accepted: 01/06/2016] [Indexed: 11/20/2022]
Abstract
Lung cancers are common malignancies, which have a very poor prognosis. These are the leading cause of cancer deaths in France and worldwide. Behind this unfavourable prognosis hides many disparities according to age, sex, social level and exposure to risk factors. The detection of the genetic abnormalities, which drive carcinogenesis has totally changed the therapeutic approach. Tumours are now classified according to their molecular profile which is itself associated with new demographic data. We here review the most recent data on this topic.
Collapse
|
774
|
Crona J, Skogseid B. GEP- NETS UPDATE: Genetics of neuroendocrine tumors. Eur J Endocrinol 2016; 174:R275-90. [PMID: 27165966 DOI: 10.1530/eje-15-0972] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022]
Abstract
Neuroendocrine tumors (NETs) are a heterogeneous group of neoplasms, arising from neuroendocrine cells that are dispersed throughout the body. Around 20% of NETs occur in the context of a genetic syndrome. Today there are at least ten recognized NET syndromes. This includes the classical syndromes: multiple endocrine neoplasias types 1 and 2, and von Hippel-Lindau and neurofibromatosis type 1. Additional susceptibility genes associated with a smaller fraction of NETs have also been identified. Recognizing genetic susceptibility has proved essential both to provide genetic counseling and to give the best preventive care. In this review we will also discuss the knowledge of somatic genetic alterations in NETs. At least 24 genes have been implicated as drivers of neuroendocrine tumorigenesis, and the overall rates of genomic instability are relatively low. Genetic intra-tumoral, as well as inter-tumoral heterogeneity in the same patient, have also been identified. Together these data point towards the common pathways in NET evolution, separating early from late disease drivers. Although knowledge of specific mutations in NETs has limited impact on actual patient management, we predict that in the near future genomic profiling of tumors will be included in the clinical arsenal for diagnostics, prognostics and therapeutic decisions.
Collapse
Affiliation(s)
- Joakim Crona
- Department of Medical SciencesUppsala University, Rudbecklaboratoriet, Dag hammarskjölds väg 20, 75185 Uppsala, Sweden
| | - Britt Skogseid
- Department of Medical SciencesUppsala University, Rudbecklaboratoriet, Dag hammarskjölds väg 20, 75185 Uppsala, Sweden
| |
Collapse
|
775
|
Castellanos EH, Horn L. Re-Evaluating Progression in an Era of Progress: A Review of First- and Second-Line Treatment Options in Anaplastic Lymphoma Kinase-Positive Non-Small Cell Lung Cancer. Oncologist 2016; 21:755-61. [PMID: 27053502 PMCID: PMC4912362 DOI: 10.1634/theoncologist.2015-0396] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/03/2016] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED : The advent of crizotinib, the first small molecule inhibitor against anaplastic lymphoma kinase (ALK), has led to impressive advances in the care of patients with advanced ALK-rearranged non-small cell lung cancer. The development of second-generation ALK inhibitors, starting with the recent U.S. Food and Drug Administration approval of ceritinib, promises to expand the therapeutic landscape for this cohort of patients. With increasing use of molecularly targeted therapy options, it has been observed that disease progression in patients receiving targeted agents has a heterogeneous biology, manifesting as either oligoprogressive or widely progressive disease, which may require development of innovative treatment strategies. This review discusses the first- and second-generation ALK inhibitors approved or in clinical development, as well as the novel challenges and approaches to disease progression in patients on targeted agents. IMPLICATIONS FOR PRACTICE The identification of driver mutations in non-small cell lung cancer (NSCLC), most prominently epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK), has expanded treatment options for a significant cohort of patients. However, the success of targeted agents has brought new challenges, particularly regarding management of progression. Progression manifests heterogeneously, and management of oligoprogression may differ from diffusely progressive disease. Multiple options for treatment at progression exist, and it is becoming evident that selecting the best avenue of care requires understanding the biology and potential drivers of disease progression. This review discusses the array of treatment options available for patients with ALK-positive NSCLC, as well as evaluation and treatment of progressive disease.
Collapse
Affiliation(s)
- Emily H Castellanos
- Department of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Leora Horn
- Department of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| |
Collapse
|
776
|
Programmed Death-1 Inhibition in Cancer With a Focus on Non-Small Cell Lung Cancer: Rationale, Nursing Implications, and Patient Management Strategies. Clin J Oncol Nurs 2016; 20:319-26. [PMID: 27206299 DOI: 10.1188/16.cjon.319-326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Programmed death-1 (PD-1) immune checkpoint inhibitors are novel immuno-oncology agents. Unlike chemotherapy or targeted agents, which inhibit tumor cell proliferation or induce tumor cell death, immune checkpoint inhibitors are designed to stimulate a patient's own immune system to eliminate tumors. As a result of their mechanism of action, PD-1 pathway inhibitors are associated with adverse events (AEs) with immunologic etiologies, termed immune-mediated AEs (imAEs). These include skin and gastrointestinal AEs, and endocrine, hepatic, renal, and respiratory AEs, including pneumonitis. Most imAEs can be effectively managed with treatment interruption/discontinuation and/or steroids or other immunosuppressive agents. A specialist consult may be required in some cases, and endocrine imAEs may require permanent hormone replacement therapy. OBJECTIVES This article provides an overview of PD-1 inhibitors, including the potential mechanism of action, key clinical trial data, and strategies for managing patients who may receive PD-1 inhibitors for the treatment of non-small cell lung cancer. METHODS Information in the article comes from PubMed literature searches and the author's experience with these agents in clinical trials. FINDINGS Oncology clinicians must thoroughly assess baseline functioning and symptoms and be vigilant for imAEs, which require prompt diagnosis and management. A good understanding of the clinical profile of PD-1 pathway inhibitors is instrumental in helping clinicians manage patients receiving these new therapies.
Collapse
|
777
|
Auliac JB. Les cancers du poumon avec une mutation oncogénique : une prise en charge de plus en plus spécifique. ONCOLOGIE 2016. [DOI: 10.1007/s10269-016-2639-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
778
|
Stein J, Mann J. Specialty pharmacy services for patients receiving oral medications for solid tumors. Am J Health Syst Pharm 2016; 73:775-96. [DOI: 10.2146/ajhp150863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jill Stein
- Department of Pharmaceutical Care, University of Iowa Health Care, Iowa City, IA
| | - Janelle Mann
- St. Louis College of Pharmacy, St. Louis, MO, and Department of Pharmacy Practice, Division of Specialty Care Pharmacy, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
779
|
Bennati C, Paglialunga L, Ricciuti B, Metro G, Marcomigni L, Gili A, Crinò L. Targeting EGFR and ALK in NSCLC: current evidence and future perspective. Lung Cancer Manag 2016; 5:79-90. [PMID: 30643552 PMCID: PMC6310340 DOI: 10.2217/lmt-2016-0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/12/2016] [Indexed: 12/26/2022] Open
Abstract
The advent of molecular therapy targeting specific driver oncogenes has dramatically changed the prognosis of a subset of NSCLC, dilating survival and improving the quality of life of patients with advanced disease. Two of the major targets for treatment with receptor TKIs are the activated mutated forms of the EGFR and the ALK gene fusions. In advanced NSCLC patients harboring EGFR mutations or ALK rearrangements, the use of TKIs in the first-line setting, have provided unexpected large progression-free survival and overall survival benefits, compared with cytotoxic chemotherapy. However, despite initial responses and durable remissions, the development of resistance inevitably leads to treatment failure. The aim of this review is to discuss the treatment strategy currently used for tumors harboring these two genetic targets and to focus on what will be available in clinical practice in the near future.
Collapse
Affiliation(s)
- Chiara Bennati
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Luca Paglialunga
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Biagio Ricciuti
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Giulio Metro
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Luca Marcomigni
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| | - Alessio Gili
- Department of Experimental Medicine, S Maria della Misericordia Hospital, Perugia/Italy
| | - Lucio Crinò
- Department of Medical Oncology, S Maria della Misericordia Hospital, Perugia/Italy
| |
Collapse
|
780
|
Croegaert K, Kolesar JM. Role of anaplastic lymphoma kinase inhibition in the treatment of non-small-cell lung cancer. Am J Health Syst Pharm 2016; 72:1456-62. [PMID: 26294238 DOI: 10.2146/ajhp140836] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Published data on the clinical efficacy, safety, dosage and administration, and costs of the anaplastic lymphoma kinase (ALK) inhibitors crizotinib and ceritinib in the treatment of non-small-cell lung cancer (NSCLC) are reviewed and compared. SUMMARY The ALK protein functions as a transmembrane receptor tyrosine kinase; rearrangements of the ALK gene are associated with the development of NSCLC with adenocarcinoma histology. Crizotinib is an oral tyrosine kinase inhibitor approved in 2011 as a first-line therapy for patients with metastatic ALK mutation-driven NSCLC. Significantly improved response rates and progression-free survival (PFS) have been reported with the use of crizotinib therapy versus standard chemotherapy, but mutations conferring resistance to treatment develop in most cases. The second-generation ALK inhibitor ceritinib was approved in 2014 for the treatment of ALK-mutated NSCLC in patients who are intolerant or develop resistance to crizotinib. In a clinical trial of ceritinib involving 130 patients with ALK-positive NSCLC, the majority of whom had experienced disease progression during crizotinib use, patients receiving at least 400 mg of ceritinib daily had an overall response rate of 56% and median PFS of seven months. Adverse effects commonly reported with the use of either drug include visual disturbances, gastrointestinal disorders (e.g., diarrhea), and liver enzyme abnormalities. CONCLUSION The tyrosine kinase inhibitors crizotinib and ceritinib provide an effective treatment approach for patients with ALK-mutated NSCLC. Efficacy data for both crizotinib and ceritinib indicate improved response rates and PFS with the use of either drug as an alternative to standard chemotherapy.
Collapse
Affiliation(s)
- Katie Croegaert
- Katie Croegaert, Pharm.D., is Postgraduate Year 2 Oncology Pharmacy Resident, University of Wisconsin (UW) Hospital and Clinics, Madison. Jill M. Kolesar, Pharm.D., BCPS, FCCP, is Professor of Pharmacy, UW School of Pharmacy, and Director, 3P Analytical Laboratory, UW Carbone Comprehensive Cancer Center, Madison
| | - Jill M Kolesar
- Katie Croegaert, Pharm.D., is Postgraduate Year 2 Oncology Pharmacy Resident, University of Wisconsin (UW) Hospital and Clinics, Madison. Jill M. Kolesar, Pharm.D., BCPS, FCCP, is Professor of Pharmacy, UW School of Pharmacy, and Director, 3P Analytical Laboratory, UW Carbone Comprehensive Cancer Center, Madison.
| |
Collapse
|
781
|
Ali SM, Hensing T, Schrock AB, Allen J, Sanford E, Gowen K, Kulkarni A, He J, Suh JH, Lipson D, Elvin JA, Yelensky R, Chalmers Z, Chmielecki J, Peled N, Klempner SJ, Firozvi K, Frampton GM, Molina JR, Menon S, Brahmer JR, MacMahon H, Nowak J, Ou SHI, Zauderer M, Ladanyi M, Zakowski M, Fischbach N, Ross JS, Stephens PJ, Miller VA, Wakelee H, Ganesan S, Salgia R. Comprehensive Genomic Profiling Identifies a Subset of Crizotinib-Responsive ALK-Rearranged Non-Small Cell Lung Cancer Not Detected by Fluorescence In Situ Hybridization. Oncologist 2016; 21:762-70. [PMID: 27245569 DOI: 10.1634/theoncologist.2015-0497] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/07/2016] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION For patients with non-small cell lung cancer (NSCLC) to benefit from ALK inhibitors, sensitive and specific detection of ALK genomic rearrangements is needed. ALK break-apart fluorescence in situ hybridization (FISH) is the U.S. Food and Drug Administration approved and standard-of-care diagnostic assay, but identification of ALK rearrangements by other methods reported in NSCLC cases that tested negative for ALK rearrangements by FISH suggests a significant false-negative rate. We report here a large series of NSCLC cases assayed by hybrid-capture-based comprehensive genomic profiling (CGP) in the course of clinical care. MATERIALS AND METHODS Hybrid-capture-based CGP using next-generation sequencing was performed in the course of clinical care of 1,070 patients with advanced lung cancer. Each tumor sample was evaluated for all classes of genomic alterations, including base-pair substitutions, insertions/deletions, copy number alterations and rearrangements, as well as fusions/rearrangements. RESULTS A total of 47 patients (4.4%) were found to harbor ALK rearrangements, of whom 41 had an EML4-ALK fusion, and 6 had other fusion partners, including 3 previously unreported rearrangement events: EIF2AK-ALK, PPM1B-ALK, and PRKAR1A-ALK. Of 41 patients harboring ALK rearrangements, 31 had prior FISH testing results available. Of these, 20 were ALK FISH positive, and 11 (35%) were ALK FISH negative. Of the latter 11 patients, 9 received crizotinib based on the CGP results, and 7 achieved a response with median duration of 17 months. CONCLUSION Comprehensive genomic profiling detected canonical ALK rearrangements and ALK rearrangements with noncanonical fusion partners in a subset of patients with NSCLC with previously negative ALK FISH results. In this series, such patients had durable responses to ALK inhibitors, comparable to historical response rates for ALK FISH-positive cases. IMPLICATIONS FOR PRACTICE Comprehensive genomic profiling (CGP) that includes hybrid capture and specific baiting of intron 19 of ALK is a highly sensitive, alternative method for identification of drug-sensitive ALK fusions in patients with non-small cell lung cancer (NSCLC) who had previously tested negative using standard ALK fluorescence in situ hybridization (FISH) diagnostic assays. Given the proven benefit of treatment with crizotinib and second-generation ALK inhibitors in patients with ALK fusions, CGP should be considered in patients with NSCLC, including those who have tested negative for other alterations, including negative results using ALK FISH testing.
Collapse
Affiliation(s)
- Siraj M Ali
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - Thomas Hensing
- Department of Medicine, North Shore University Health System, Evanston, Illinois, USA Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | - Justin Allen
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - Eric Sanford
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - Kyle Gowen
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - Atul Kulkarni
- Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Jie He
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - James H Suh
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - Doron Lipson
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | - Julia A Elvin
- Foundation Medicine Inc., Cambridge, Massachusetts, USA
| | | | | | | | - Nir Peled
- Davidoff Cancer Center, Tiqwa, Israel
| | - Samuel J Klempner
- Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Orange, California, USA
| | | | | | | | - Smitha Menon
- Froedtert Cancer Center, Milwaukee, Wisconsin, USA
| | - Julie R Brahmer
- Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Heber MacMahon
- Department of Radiology, The University of Chicago, Chicago, Illinois, USA
| | - Jan Nowak
- Department of Pathology, North Shore University Health System, Evanston, Illinois, USA
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, School of Medicine, University of California, Irvine, Orange, California, USA
| | | | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, Manhattan, New York, USA
| | - Maureen Zakowski
- Memorial Sloan Kettering Cancer Center, Manhattan, New York, USA
| | | | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, Massachusetts, USA Albany Medical College, Albany, New York, USA
| | | | | | - Heather Wakelee
- Department of Medicine, Division of Oncology, School of Medicine, Stanford University, Stanford, California, USA
| | - Shridar Ganesan
- Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Ravi Salgia
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
782
|
Salgia R. Mutation testing for directing upfront targeted therapy and post-progression combination therapy strategies in lung adenocarcinoma. Expert Rev Mol Diagn 2016; 16:737-49. [PMID: 27139190 PMCID: PMC4926789 DOI: 10.1080/14737159.2016.1181545] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Advances in the biology of non-small-cell lung cancer, especially adenocarcinoma, reveal multiple molecular subtypes driving oncogenesis. Accordingly, individualized targeted therapeutics are based on mutational diagnostics. Areas covered: Advances in strategies and techniques for individualized treatment, particularly of adenocarcinoma, are described through literature review. Approved therapies are established for some molecular subsets, with new driver mutations emerging that represent increasing proportions of patients. Actionable mutations are denovo oncogenic drivers or acquired resistance mediators, and mutational profiling is important for directing therapy. Patients should be monitored for emerging actionable resistance mutations. Liquid biopsy and associated multiplex diagnostics will be important means to monitor patients during treatment. Expert commentary: Outcomes with targeted agents may be improved by integrating mutation screens during treatment to optimize subsequent therapy. In order for this to be translated into impactful patient benefit, appropriate platforms and strategies need to be optimized and then implemented universally.
Collapse
Affiliation(s)
- Ravi Salgia
- a Department of Medical Oncology and Therapeutics Research , City of Hope , Duarte , CA , USA
| |
Collapse
|
783
|
Targeted next generation sequencing of parotid gland cancer uncovers genetic heterogeneity. Oncotarget 2016; 6:18224-37. [PMID: 26053092 PMCID: PMC4627247 DOI: 10.18632/oncotarget.4015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/14/2015] [Indexed: 12/12/2022] Open
Abstract
Salivary gland cancer represents a heterogeneous group of malignant tumors. Due to their low incidence and the existence of multiple morphologically defined subtypes, these tumors are still poorly understood with regard to their molecular pathogenesis and therapeutically relevant genetic alterations. Performing a systematic and comprehensive study covering 13 subtypes of salivary gland cancer, next generation sequencing was done on 84 tissue samples of parotid gland cancer using multiplex PCR for enrichment of cancer related gene loci covering hotspots of 46 cancer genes. Mutations were identified in 22 different genes. The most frequent alterations affected TP53, followed by RAS genes, PIK3CA, SMAD4 and members of the ERB family. HRAS mutations accounted for more than 90% of RAS mutations, occurring especially in epithelial-myoepithelial carcinomas and salivary duct carcinomas. Additional mutations in PIK3CA also affected particularly epithelial-myoepithelial carcinomas and salivary duct carcinomas, occurring simultaneously with HRAS mutations in almost all cases, pointing to an unknown and therapeutically relevant molecular constellation. Interestingly, 14% of tumors revealed mutations in surface growth factor receptor genes including ALK, HER2, ERBB4, FGFR, cMET and RET, which might prove to be targetable by new therapeutic agents. 6% of tumors revealed mutations in SMAD4. In summary, our data provide novel insight into the fundamental molecular heterogeneity of salivary gland cancer, relevant in terms of tumor classification and the establishment of targeted therapeutic concepts.
Collapse
|
784
|
Abstract
INTRODUCTION Lung cancer is associated with poor prognosis and limited benefit from chemotherapy. The treatment of non-small cell lung cancer (NSCLC) has been revolutionized by the discovery of targetable genetic alterations, including the ALK fusion oncogene. AREAS COVERED Three drugs have been approved for clinical use in ALK-positive patients - crizotinib, ceritinib and alectinib. Unfortunately, treatment resistance inevitably develops. Several mechanisms of acquired resistance are reported. In this review, we will discuss emerging treatment options in ALK-positive advanced NSCLC and strategies to overcome resistance mechanisms, including newer generation of ALK inhibitors, Hsp90 inhibitors and immunotherapy. EXPERT OPINION Tremendous advances have been made in the treatment of ALK-positive lung cancers, but management hurdles still exist, including universal development of resistance to ALK inhibitors and limited CNS activity. Given that specific treatment strategies target distinct patterns of resistance, re-biopsy at the time of progression appears necessary to optimize management. However, there remain many issues in routine clinical application including the burden placed on the patients by serial biopsies and the risks of repeat invasive procedures. Future studies are needed to validate the usage of non- or minimally invasive tests and to determine the optimal orders of utilizing different ALK inhibitors.
Collapse
Affiliation(s)
- Janaki Sharma
- a Division of Medical Oncology , Montefiore Medical Center, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Vipul Pareek
- a Division of Medical Oncology , Montefiore Medical Center, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Huijie Liu
- a Division of Medical Oncology , Montefiore Medical Center, Albert Einstein College of Medicine , Bronx , NY , USA
| | - Haiying Cheng
- a Division of Medical Oncology , Montefiore Medical Center, Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
785
|
Helsten T, Kato S, Schwaederle M, Tomson BN, Buys TPH, Elkin SK, Carter JL, Kurzrock R. Cell-Cycle Gene Alterations in 4,864 Tumors Analyzed by Next-Generation Sequencing: Implications for Targeted Therapeutics. Mol Cancer Ther 2016; 15:1682-90. [PMID: 27196769 DOI: 10.1158/1535-7163.mct-16-0071] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/29/2016] [Indexed: 11/16/2022]
Abstract
Alterations in the cyclin-dependent kinase (CDK)-retinoblastoma (RB) machinery disrupt cell-cycle regulation and are being targeted in drug development. To understand the cancer types impacted by this pathway, we analyzed frequency of abnormalities in key cell-cycle genes across 4,864 tumors using next-generation sequencing (182 or 236 genes; Clinical Laboratory Improvement Amendments laboratory). Aberrations in the cell-cycle pathway were identified in 39% of cancers, making this pathway one of the most commonly altered in cancer. The frequency of aberrations was as follows: CDKN2A/B (20.1% of all patients), RB1 (7.6%), CCND1 (6.1%), CCNE1 (3.6%), CDK4 (3.2%), CCND3 (1.8%), CCND2 (1.7%), and CDK6 (1.7%). Rates and types of aberrant cell-cycle pathway genes differed between cancer types and within histologies. Analysis of coexisting and mutually exclusive genetic aberrations showed that CCND1, CCND2, and CCND3 aberrations were all positively associated with CDK6 aberrations [OR and P values, multivariate analysis: CCND1 and CDK6 (OR = 3.5; P < 0.0001), CCND2 and CDK6 (OR = 4.3; P = 0.003), CCND3 and CDK6 (OR = 3.6; P = 0.007)]. In contrast, RB1 alterations were negatively associated with multiple gene anomalies in the cell-cycle pathway, including CCND1 (OR = 0.25; P = 0.003), CKD4 (OR = 0.10; P = 0.001), and CDKN2A/B (OR = 0.21; P < 0.0001). In conclusion, aberrations in the cell-cycle pathway were very common in diverse cancers (39% of 4,864 neoplasms). The frequencies and types of alterations differed between and within tumor types and will be informative for drug development strategies. Mol Cancer Ther; 15(7); 1682-90. ©2016 AACR.
Collapse
Affiliation(s)
- Teresa Helsten
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, La Jolla, California
| | - Shumei Kato
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Maria Schwaederle
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, La Jolla, California
| | | | | | | | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, UC San Diego Moores Cancer Center, La Jolla, California
| |
Collapse
|
786
|
EGFR-Mediated Reactivation of MAPK Signaling Induces Acquired Resistance to GSK2118436 in BRAF V600E–Mutant NSCLC Cell Lines. Mol Cancer Ther 2016; 15:1627-36. [DOI: 10.1158/1535-7163.mct-15-0375] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 04/23/2016] [Indexed: 11/16/2022]
|
787
|
Russo A, Franchina T, Ricciardi GRR, Ferraro G, Scimone A, Bronte G, Russo A, Rolfo C, Adamo V. Central nervous system involvement in ALK-rearranged NSCLC: promising strategies to overcome crizotinib resistance. Expert Rev Anticancer Ther 2016; 16:615-23. [PMID: 27109446 DOI: 10.1080/14737140.2016.1182427] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION ALK rearranged Non Small Cell Lung Cancers (NSCLCs) represent a distinct subgroup of patients with peculiar clinic-pathological features. These patients exhibit dramatic responses when treated with the ALK tyrosine kinase inhibitor Crizotinib, albeit Central Nervous System (CNS) activity is much less impressive than that observed against extracranial lesions. CNS involvement has become increasingly observed in these patients, given their prolonged survival. Several novel generation ALK inhibitors have been developing to increase CNS penetration and to provide more complete ALK inhibition.. AREAS COVERED The CNS activity of Crizotinib and novel generation ALK inhibitors will be summarized in this review, evaluating the strengths and weaknesses of the therapeutic strategies developed to date in this specific subgroup of NSCLCs with a look towards the future. Expert commentary: In the next few years, the results of ongoing comparative head-to-head trials will provide the definitive conclusions on the optimal treatment sequence in ALK-rearranged NSCLCs. Moreover, ongoing clinical trials with novel-generation ALK inhibitors will produce more evidences on the best approach in the growing number of ALK-positive NSCLCs with CNS involvement.
Collapse
Affiliation(s)
- Alessandro Russo
- a Medical Oncology Unit A.O. Papardo and Department of Human Pathology , University of Messina , Messina , Italy
| | - Tindara Franchina
- a Medical Oncology Unit A.O. Papardo and Department of Human Pathology , University of Messina , Messina , Italy
| | | | - Giuseppa Ferraro
- a Medical Oncology Unit A.O. Papardo and Department of Human Pathology , University of Messina , Messina , Italy
| | - Antonino Scimone
- a Medical Oncology Unit A.O. Papardo and Department of Human Pathology , University of Messina , Messina , Italy
| | - Giuseppe Bronte
- b Department of Surgical, Oncological and Oral Sciences , University of Palermo , Palermo , Italy
| | - Antonio Russo
- b Department of Surgical, Oncological and Oral Sciences , University of Palermo , Palermo , Italy
| | - Christian Rolfo
- c Department of Medical Oncology , Antwerp University Hospital , Antwerp , Belgium
| | - Vincenzo Adamo
- a Medical Oncology Unit A.O. Papardo and Department of Human Pathology , University of Messina , Messina , Italy
| |
Collapse
|
788
|
Ha D, Mazzone PJ, Ries AL, Malhotra A, Fuster M. The Utility of Exercise Testing in Patients with Lung Cancer. J Thorac Oncol 2016; 11:1397-410. [PMID: 27156441 DOI: 10.1016/j.jtho.2016.04.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 12/25/2022]
Abstract
The harm associated with lung cancer treatment include perioperative morbidity and mortality and therapy-induced toxicities in various organs, including the heart and lungs. Optimal treatment therefore entails a need for risk assessment to weigh the probabilities of benefits versus harm. Exercise testing offers an opportunity to evaluate a patient's physical fitness/exercise capacity objectively. In lung cancer, it is most often used to risk-stratify patients undergoing evaluation for lung cancer resection. In recent years, its use outside this context has been described, including in nonsurgical candidates and lung cancer survivors. In this article we review the physiology of exercise testing and lung cancer. Then, we assess the utility of exercise testing in patients with lung cancer in four contexts (preoperative evaluation for lung cancer resection, after lung cancer resection, lung cancer prognosis, and assessment of efficiency of exercise training programs) after systematically identifying original studies involving the most common forms of exercise tests in this patient population: laboratory cardiopulmonary exercise testing and simple field testing with the 6-minute walk test, shuttle walk test, and/or stair-climbing test. Lastly, we propose a conceptual framework for risk assessment of patients with lung cancer who are being considered for therapy and identify areas for further studies in this patient population.
Collapse
Affiliation(s)
- Duc Ha
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, California.
| | | | - Andrew L Ries
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, California
| | - Atul Malhotra
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, California
| | - Mark Fuster
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, California; Pulmonary and Critical Care Medicine Section, Veterans Administration San Diego Healthcare System, San Diego, California
| |
Collapse
|
789
|
Phase I Study of Ceritinib (LDK378) in Japanese Patients with Advanced, Anaplastic Lymphoma Kinase-Rearranged Non-Small-Cell Lung Cancer or Other Tumors. J Thorac Oncol 2016; 10:1058-66. [PMID: 26020125 PMCID: PMC4467585 DOI: 10.1097/jto.0000000000000566] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Introduction: Anaplastic lymphoma kinase (ALK)-rearranged non–small-cell lung cancer (NSCLC) is sensitive to ALK inhibitors, but resistance develops. This study assessed the maximum-tolerated dose, safety, pharmacokinetics (PK), and antitumor activity of ceritinib, a novel ALK inhibitor (ALKi), in Japanese patients with ALK-rearranged malignancies. Methods: This phase I, multicenter, open-label study (NCT01634763) enrolled adult patients with ALK-rearranged (by fluorescence in situ hybridization and/or immunohistochemistry) locally advanced/metastatic malignancy that had progressed despite standard therapy. The study comprised two parts: dose escalation and dose expansion. Ceritinib (single-dose) was administered orally in the 3-day PK run-in period, then once daily, in 21-day cycles. Adaptive dose escalations were guided by a Bayesian model. Results: Twenty patients (80% with ALKi treatment history [ALKi-pretreated]; 19 NSCLC; one inflammatory myofibroblastic tumor) received ceritinib 300 to 750 mg (19 during dose escalation, one in dose expansion). Two dose-limiting toxicities occurred: grade 3 lipase increase (600 mg); grade 3 drug-induced liver injury (750 mg). The most common adverse events were gastrointestinal (nausea: 95%; diarrhea, vomiting: 75%). Ceritinib PK profile was dose proportional across 300 to 750 mg dosages; steady state was reached by day 15. Overall response rate was 55% (11 of 20 patients). Among patients with NSCLC, partial response was observed in two of four ALKi-naive patients, five of nine crizotinib-pretreated patients, two of four alectinib-pretreated patients, and one of two crizotinib and alectinib/ASP3026 pretreated patients. The ASP3026-pretreated inflammatory myofibroblastic tumor patient achieved partial response. Conclusions: Ceritinib maximum-tolerated dose was 750 mg once daily in Japanese patients. Antitumor activity was observed irrespective of prior ALKi treatment history. Dose expansion, examining the activity of ceritinib in alectinib-resistant patients, is ongoing.
Collapse
|
790
|
Rothschild SI. New treatment options for ALK+ advanced non-small-cell lung cancer: critical appraisal of ceritinib. Ther Clin Risk Manag 2016; 12:735-41. [PMID: 27217763 PMCID: PMC4863587 DOI: 10.2147/tcrm.s87876] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Rearrangements in ALK gene and EML4 gene were first described in 2007. This genomic aberration is found in about 2%–8% of non-small-cell lung cancer (NSCLC) patients. Crizotinib was the first ALK tyrosine kinase inhibitor licensed for the treatment of metastatic ALK-positive NSCLC based on a randomized Phase III trial. Despite the initial treatment response of crizotinib, disease progression inevitably develops after approximately 10 months of therapy. Different resistance mechanisms have recently been described. One relevant mechanism of resistance is the development of mutations in ALK. Novel ALK tyrosine kinase inhibitors have been developed to overcome these mutations. Ceritinib is an oral second-generation ALK inhibitor showing clinical activity not only in crizotinib-resistant ALK-positive NSCLC but also in treatment-naïve ALK-positive disease. In this paper, preclinical and clinical data of ceritinib are reviewed, and its role in the clinical setting is put into perspective.
Collapse
Affiliation(s)
- Sacha I Rothschild
- Department of Internal Medicine, Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
791
|
Mussat E, Giraud V, Julie C, Chinet T, Leprieur EG. Fatal Haemoptysis Associated with Dramatic Response to Crizotinib in an ALK-Rearranged Lung Adenocarcinoma. J Clin Diagn Res 2016; 10:XD01-XD03. [PMID: 27134984 DOI: 10.7860/jcdr/2016/17805.7428] [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: 11/12/2015] [Accepted: 01/09/2016] [Indexed: 11/24/2022]
Abstract
The presence of an ALK (Anaplastic Lymphoma Kinase) rearrangement is a rare molecular feature in Non-Small Cell Lung Carcinoma (NSCLC), and concerns mainly non- or light smokers, young patients, with adenocarcinoma histological type. These tumours are particularly sensitive to Alk-targeted therapies, as crizotinib. Crizotinib is usually well-tolerated. We report a case of fatal haemoptysis associated with dramatic response to crizotinib in a patient with an ALK-rearranged lung adenocarcinoma. The patient presented a mediastinal invasion with tracheal involvement and compression of the right pulmonary artery. The initial evolution under crizotinib was good with tumour response. At 6 weeks of crizotinib the patient presented a massive haemoptysis with a tracheobronchial fistula and pneumomediastinum. She died of acute respiratory failure. Our case is the first to report a fatal effect of crizotinib associated with tumour necrosis and good tumour response on a massive mediastinal infiltration. Precautions are recommended with the use of crizotinib in proximal lung tumours with vascular invasion.
Collapse
Affiliation(s)
- Elodie Mussat
- Faculty, Department of Respiratory Diseases and Thoracic Oncology, Ambroise Pare Hospital - APHP , Boulogne-Billancourt, France
| | - Violaine Giraud
- Faculty, Department of Respiratory Diseases and Thoracic Oncology, Ambroise Pare Hospital - APHP , Boulogne-Billancourt, France
| | - Catherine Julie
- Faculty, Department of Pathology, Ambroise Pare Hospital - APHP , Boulogne-Billancourt, France
| | - Thierry Chinet
- Faculty, Department of Respiratory Diseases and Thoracic Oncology, Ambroise Pare Hospital - APHP , Boulogne-Billancourt, France. EA 4340 BCOH, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
| | - Etienne Giroux Leprieur
- Faculty, Department of Respiratory Diseases and Thoracic Oncology, Ambroise Pare Hospital - APHP , Boulogne-Billancourt, France. EA 4340 BCOH, UVSQ, Paris-Saclay University, Boulogne-Billancourt, France
| |
Collapse
|
792
|
Kerner GSMA, Koole MJB, Bongaerts AHH, Pruim J, Groen HJM, CTMM Air Force Consortium. Total Body Metabolic Tumor Response in ALK Positive Non-Small Cell Lung Cancer Patients Treated with ALK Inhibition. PLoS One 2016; 11:e0149955. [PMID: 27137772 PMCID: PMC4854442 DOI: 10.1371/journal.pone.0149955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/15/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND In ALK-positive advanced NSCLC, crizotinib has a high response rate and effectively increases quality of life and survival. CT measurement of the tumor may insufficiently reflect the actual tumor load changes during targeted therapy with crizotinib. We explored whether 18F-FDG PET measured metabolic changes are different from CT based changes and studied the impact of these changes on disease progression. METHODS 18F-FDG PET/CT was performed prior to and after 6 weeks of crizotinib treatment. Tumor response on CT was classified with RECIST 1.1, while 18F-FDG PET response was assessed according to the 1999 EORTC recommendations and PERCIST criteria. Agreement was assessed using McNemars test. During follow-up, patients received additional PET/CT during crizotinib treatment and second generation ALK inhibition. We assessed whether PET was able to detect progression earlier then CT. RESULTS In this exploratory study 15 patients were analyzed who were treated with crizotinib. There was a good agreement in the applicability of CT and 18F-FDG PET/CT using the EORTC recommendations. During first line crizotinib and subsequent second line ALK inhibitors, PET was able to detect progression earlier then CT in 10/22 (45%) events of progression and in the others disease progression was detected simultaneously. CONCLUSION In advanced ALK positive NSCLC PET was able to detect progressive disease earlier than with CT in nearly half of the assessments while both imaging tests performed similar in the others.
Collapse
Affiliation(s)
- Gerald S. M. A. Kerner
- University of Groningen and Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, the Netherlands
- * E-mail:
| | - Michel J. B. Koole
- University of Groningen and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, the Netherlands
| | - Alphons H. H. Bongaerts
- University of Groningen and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Pruim
- University of Groningen and Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, the Netherlands
- Department of Nuclear Medicine, Tygerberg Hospital, Stellenbosch University, Stellenbosch, South-Africa
| | - Harry J. M. Groen
- University of Groningen and Department of Pulmonary Diseases, University Medical Center Groningen, Groningen, the Netherlands
| | | |
Collapse
|
793
|
Chiang NJ, Hsu C, Chen JS, Tsou HH, Shen YY, Chao Y, Chen MH, Yeh TS, Shan YS, Huang SF, Chen LT. Expression levels of ROS1/ALK/c-MET and therapeutic efficacy of cetuximab plus chemotherapy in advanced biliary tract cancer. Sci Rep 2016; 6:25369. [PMID: 27136744 PMCID: PMC4853728 DOI: 10.1038/srep25369] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/15/2016] [Indexed: 12/26/2022] Open
Abstract
Aberrant expression of ROS1, ALK or c-MET (RAM) is implicated in carcinogenesis and cancer drug resistance. We retrospectively evaluated the effect of RAM expression on outcomes for advanced biliary tract cancer patients, who were treated with gemcitabine plus oxaliplatin (GEMOX), with or without cetuximab, in a randomized phase II trial. RAM expression levels on archived tissue sections were scored using immunohistochemistry (IHC). Of 110 tumors with IHC staining for all three markers, 18 were RAMhigh (IHC intensity 3+ for any markers). Ninety-two tumors were RAMlow (IHC intensity <3+ for all markers). All RAMhigh tumors were intra-hepatic cholangiocarcinomas (IHCC). Of the patients with IHCC (n = 80), median overall survival (OS) of RAMhigh group was inferior to that of the RAMlow group (5.7 vs. 11.7 months, p = 0.021). In multivariate analysis RAMhigh remained an independently adverse prognostic factor, with a hazard ratio of 2.01 (p = 0.039). In the RAMlow group, GEMOX treatment with cetuximab significantly improved the disease control rate (68% vs. 41%, p = 0.044), median progression-free survival (7.3 vs. 4.9 months, p = 0.026), and marginally prolonged median OS (14.1 vs 9.6 months, p = 0.056), compared to GEMOX treatment alone. Future trials of anti-EGFR inhibitors for IHCC may consider RAM expression as a patient stratification factor.
Collapse
Affiliation(s)
- Nai-Jung Chiang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, and National Taiwan University Cancer Center, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jen-Shi Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Hui Tsou
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan.,Graduate Institute of Biostatistics, College of Public Health, China Medical University, Taichung, Taiwan
| | - Ying-Ying Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Huang Chen
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Institute of Clinical Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
794
|
Brustugun OT. Stratification in advanced non-small cell lung cancer: precision medicine in practice. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1176861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
795
|
A causal link from ALK to hexokinase II overexpression and hyperactive glycolysis in EML4-ALK-positive lung cancer. Oncogene 2016; 35:6132-6142. [PMID: 27132509 PMCID: PMC5093092 DOI: 10.1038/onc.2016.150] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/05/2016] [Accepted: 03/24/2016] [Indexed: 12/19/2022]
Abstract
A high rate of aerobic glycolysis is a hallmark of malignant transformation. Accumulating evidence suggests that diverse regulatory mechanisms mediate this cancer-associated metabolic change seen in a wide spectrum of cancer. The echinoderm microtubule associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) fusion protein is found in approximately 3-7% of non-small cell lung carcinomas (NSCLC). Molecular evidence and therapeutic effectiveness of FDA-approved ALK inhibitors indicated that EML4-ALK is a driving factor of lung tumorigenesis. A recent clinical study showed that NSCLC harboring EML4-ALK rearrangements displayed higher glucose metabolism compared to EML4-ALK-negative NSCLC. In the current work, we presented evidence that EML4-ALK is coupled to overexpression of hexokinase II (HK2), one of the rate-limiting enzymes of the glycolytic pathway. The link from EML4-ALK to HK2 upregulation is essential for a high rate of glycolysis and proliferation of EML4-ALK-rearranged NSCLC cells. We identified hypoxia-inducible factor 1α (HIF1α) as a key transcription factor to drive HK2 gene expression in normoxia in these cells. EML4-ALK induced hypoxia-independent but glucose-dependent accumulation of HIF1α protein via both transcriptional activation of HIF1α mRNA and the PI3K-AKT pathway to enhance HIF1α protein synthesis. The EML4-ALK-mediated upregulation of HIF1α, HK2 and glycolytic metabolism was also highly active in vivo as demonstrated by FDG-PET imaging of xenografts grown from EML4-ALK-positive NSCLC cells. Our data reveal a novel EML4-ALK-HIF1α-HK2 cascade to enhance glucose metabolism in EML4-ALK-positive NSCLC.
Collapse
|
796
|
Complete and Repeated Response of a Metastatic ALK-rearranged Inflammatory Myofibroblastic Tumor to Crizotinib in a Teenage Girl. J Pediatr Hematol Oncol 2016; 38:308-11. [PMID: 26808369 DOI: 10.1097/mph.0000000000000498] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Inflammatory myofibroblastic tumors (IMT) are rare tumors in children and young adults, considered by the World Health Organization to be intermediate malignancies and rarely metastasizing, with the presence of an anaplastic lymphoma kinase rearrangement in about 50% of the cases. We report the case of a teenager who presented with a metastatic aggressive IMT that was life-threatening despite multiple treatments, and which responded repeatedly to anaplastic lymphoma kinase-targeted crizotinib therapy. Crizotinib induced drastic primary tumor regression, which was sufficient to allow surgical resection and to control distant disease. This case shows that crizotinib is a promising therapy in IMT, even in adolescents and young adults.
Collapse
|
797
|
Synthetic approaches to the 2014 new drugs. Bioorg Med Chem 2016; 24:1937-80. [DOI: 10.1016/j.bmc.2016.03.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 12/31/2022]
|
798
|
Romanidou O, Landi L, Cappuzzo F, Califano R. Overcoming resistance to first/second generation epidermal growth factor receptor tyrosine kinase inhibitors and ALK inhibitors in oncogene-addicted advanced non-small cell lung cancer. Ther Adv Med Oncol 2016; 8:176-87. [PMID: 27239236 PMCID: PMC4872250 DOI: 10.1177/1758834016631531] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) activating mutations and anaplastic lymphoma kinase (ALK) gene rearrangement in advanced non-small cell lung cancer (NSCLC) represent the two oncogenic events with an impact on current clinical practice. EGFR tyrosine kinase inhibitors (TKIs) and crizotinib are the standard of care for the treatment of EGFR mutant and ALK gene rearranged advanced NSCLC patients. Unfortunately, despite initial clinical benefit, acquired resistance to EGFR-TKIs or crizotinib usually develops after an average of 10-12 months of treatment. The aim of this review is to describe the mechanisms of resistance to first/second generation EGFR-TKIs and crizotinib. In particular, we focus on strategies to overcome resistance due to secondary EGFR T790M mutation and mutations of the ALK domain.
Collapse
Affiliation(s)
- Ourania Romanidou
- Cancer Research UK Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK, and Medical Oncology Unit, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Lorenza Landi
- Department of Medical Oncology, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Federico Cappuzzo
- Department of Medical Oncology, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Raffaele Califano
- Cancer Research UK Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
799
|
Ceritinib in patients with advanced anaplastic lymphoma kinase-rearranged anaplastic large-cell lymphoma. Blood 2016; 126:1257-8. [PMID: 26337354 DOI: 10.1182/blood-2014-12-617779] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
800
|
Kurzrock R, Giles FJ. Precision oncology for patients with advanced cancer: the challenges of malignant snowflakes. Cell Cycle 2016; 14:2219-21. [PMID: 26030337 PMCID: PMC4615125 DOI: 10.1080/15384101.2015.1041695] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Precision oncology implies customizing treatment to the unique molecular and biologic characteristics of each individual and their cancer. Its implementation is being facilitated by remarkable technological advances in genomic sequencing, as well as the increasing availability of targeted and immunotherapeutic drugs. Yet, next generation sequencing may be a disruptive technology in that its results suggest that classic paradigms for clinical research and practice are a poor fit with the complex reality encountered in metastatic malignancies. Indeed, it is evident that advanced tumors have heterogeneous molecular landscapes that mostly differ between patients. Traditional modes of clinical research/practice are drug centered, with a strategy of finding commonalities between patients so that they can be grouped together and treated similarly. However, if each patient with metastatic cancer has a unique molecular portfolio, a new patient-centered, N-of-one approach that utilizes individually tailored treatment is needed.
Collapse
Affiliation(s)
- Razelle Kurzrock
- a Center for Personalized Cancer Therapy; Division of Hematology and Oncology; UC San Diego Moores Cancer Center ; San Diego , CA USA
| | | |
Collapse
|