1
|
Michaels L, Noor M, Aslam T. Clinical and imaging strategies for the assessment of the ocular side effects of systemic targeted anti-cancer therapies. Eur J Cancer 2025; 222:115452. [PMID: 40306116 DOI: 10.1016/j.ejca.2025.115452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Systemic targeted anti-cancer therapies selectively target cancerous cells whilst limiting systemic side effects. The eye however, is a particularly sensitive organ and the expanding use of the newer targeted chemotherapy agents has been associated with multiple ocular side effects. In this review we provide an update of the ocular side effects of the newer targeted chemotherapy agents along with suggested minimum, pragmatic, evidence-based strategies for effective screening or monitoring for potential ocular side effects. This framework is designed to guide oncologists, trial managers, protocol developers and regulatory authorities so that appropriate ophthalmic clinical examinations and non-invasive modern imaging can be requested and commissioned according to a patient's specific treatment.
Collapse
Affiliation(s)
- Luke Michaels
- St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, United Kingdom
| | - Maha Noor
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom
| | - Tariq Aslam
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom; School of Health Sciences, University of Manchester, Oxford Road, Manchester M139PL, United Kingdom.
| |
Collapse
|
2
|
Baruah V, Krishna BA, Kelly MC, Qi X, O'Connor CM. Inhibition of MAPK signaling suppresses cytomegalovirus reactivation in CD34 + Kasumi-3 cells. Antiviral Res 2025; 238:106169. [PMID: 40250631 DOI: 10.1016/j.antiviral.2025.106169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Reactivation of latent human cytomegalovirus (CMV) can lead to severe complications in individuals with dysregulated immune systems. While antiviral therapies for CMV are approved, these compounds are limited by their toxicity and inability to specifically target the latent reservoir or prevent reactivation. Herein we show that CMV reactivation in Kasumi-3 cells, a CD34+ hematopoietic cell line, requires mitogen-activated protein kinase (MAPK) activation. Importantly, pharmacological inhibition of the MAPK signaling pathway, including MEK and ERK, restricts viral reactivation in Kasumi-3 cells. In sum, our findings show MEK-ERK signaling is critical for CMV reactivation, revealing a potential avenue for therapeutic intervention to prevent viral reactivation and downstream pathogenesis that is often detrimental for immunosuppressed and immunocompromised patients.
Collapse
Affiliation(s)
- Vargab Baruah
- Infection Biology, Sheikha Fatima Bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| | - Benjamin A Krishna
- Infection Biology, Sheikha Fatima Bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| | - Michael C Kelly
- Infection Biology, Sheikha Fatima Bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| | - Xu Qi
- Infection Biology, Sheikha Fatima Bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| | - Christine M O'Connor
- Infection Biology, Sheikha Fatima Bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
3
|
Kwon YJ, Kim DY, Kim UI, Kim SH, Kim YH, Kim K, Kim JS. Basroparib overcomes acquired resistance to MEK inhibitors by inhibiting Wnt-mediated cancer stemness in KRAS-mutated colorectal cancer. Biochem Pharmacol 2025; 235:116842. [PMID: 40024348 DOI: 10.1016/j.bcp.2025.116842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/07/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Mitogen-activated protein kinase (MEK) inhibitors show promise in treating KRAS-mutated cancers, including colorectal cancer (CRC). However, acquired resistance to MEK inhibitors often results in failure of effective treatment in patients with KRAS-mutated CRC. Here, we demonstrated that basroparib overcomes MEK inhibitor resistance in KRAS-G12V- or -G12D-mutated CRC. This basroparib-mediated sensitization was dependent on the KRAS mutation status and was enhanced specifically in KRAS-G12V- or -G12D-mutated cells. Additionally, when combined with MEK inhibitors, basroparib significantly reduced tumor growth in KRAS-G12V-mutated CRC xenograft models, but not in KRAS-G13D-mutated and -wild-type models. Furthermore, basroparib sensitized KRAS-G12V-mutated CRC cells with acquired resistance to MEK inhibitors. Notably, when combined with MEK inhibitors, basroparib not only suppressed tumor regrowth but also prolonged the survival of tumor models with acquired resistance to MEK inhibitors. Mechanistically, basroparib suppressed Wnt-mediated cancer stemness, a bypass mechanism for acquired resistance to MEK inhibitors in KRAS-mutated CRC. This study provides the first preclinical evidence of the relevance of basroparib in treating CRC with acquired resistance to MEK inhibitors due to APC and KRAS mutations, possibly by reducing the Wnt-mediated cancer stemness.
Collapse
Affiliation(s)
- Young-Ju Kwon
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul, Republic of Korea
| | - Dong Young Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Uk-Il Kim
- ST Pharm Co., Ltd., Seoul, Republic of Korea
| | | | - Ye-Hyun Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul, Republic of Korea
| | | | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Radiological and Medico-Oncological Sciences, University of Science and Technology, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Ma YT, Li C, Shen Y, You WH, Han MX, Mu YF, Han FJ. Mechanisms of the JNK/p38 MAPK signaling pathway in drug resistance in ovarian cancer. Front Oncol 2025; 15:1533352. [PMID: 40352594 PMCID: PMC12063130 DOI: 10.3389/fonc.2025.1533352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal malignancy in the female reproductive system, and chemotherapy drug resistance is the main cause of treatment failure. The Mitogen-Activated Protein Kinases (MAPK) pathway plays a pivotal role in regulating cell proliferation, migration, and invasive capacity in response to extracellular stimuli. This review focuses on the mechanisms and therapeutic strategies related to the JNK/p38 MAPK signaling pathway in OC resistance. The JNK/p38 MAPK pathway plays a dual role in OC chemoresistance. This review examines its role in mediating OC treatment resistance by exploring the mechanisms of action of the JNK/p38 MAPK signaling pathway, particularly its involvement in several key biological processes, including apoptosis, autophagy, DNA damage response, the tumor microenvironment (TME), and drug efflux. Additionally, the review investigates the timing of activation of this pathway and its crosstalk with other signaling pathways such as PI3K/AKT and NF-κB. Targeting JNK/p38 MAPK signaling has shown promise in reversing chemoresistance, with several inhibitors and natural compounds demonstrating potential in preclinical studies. Regulating JNK/p38 MAPK may transform what was once a terminal obstacle into a manageable challenge for OC patients with chemotherapy resistance, ultimately improving survival and quality of life.
Collapse
Affiliation(s)
- Yu-Ting Ma
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Chan Li
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ying Shen
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Wan-Hui You
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ming-Xuan Han
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yi-Fan Mu
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Feng-Juan Han
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
Aroldi F, Elez E, André T, Perkins G, Prenen H, Popovici V, Gallagher P, Houlden J, Collins L, Roberts C, Rolfo C, Di Nicolantonio F, Grayson M, Boyd R, Bettens K, Delfavero J, Coyle V, Lawler M, Khawaja H, Laurent-Puig P, Salto-Tellez M, Maughan TS, Tabernero J, Adams R, Jones R, Hennessy BT, Bardelli A, Peeters M, Middleton MR, Wilson RH, Van Schaeybroeck S. A Phase Ia/b study of MEK1/2 inhibitor binimetinib with MET inhibitor crizotinib in patients with RAS mutant advanced colorectal cancer (MErCuRIC). BMC Cancer 2025; 25:658. [PMID: 40211189 PMCID: PMC11984268 DOI: 10.1186/s12885-025-14068-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Targeting RAS mutant (MT) colorectal cancer (CRC) remains a difficult challenge, mainly due to the pervasiveness of RAS/MEK-mediated feedback loops. Preclinical studies identified MET/STAT3 as an important mediator of resistance to KRAS-MEK1/2 blockade in RASMT CRC. This dose escalation/expansion study assessed safety and initial efficacy of the MEK1/2 inhibitor binimetinib with MET inhibitor crizotinib in RASMT advanced CRC patients. METHODS In the dose escalation phase, patients with advanced solid tumours received binimetinib with crizotinib, using a rolling- 6 design to determine the maximum tolerable dose (MTD) and safety/tolerability. A subsequent dose expansion in RASMT CRC patients assessed treatment response. Blood samples for pharmacokinetics, MET biomarker and ctDNA analyses, and skin/tumour biopsies for pharmacodynamics, c-MET immunohistochemistry (IHC), MET in situ hybridisation (ISH) and MET DNA-ISH analyses were collected. RESULTS Twenty patients were recruited in 3 cohorts in the dose escalation. The MTD was binimetinib 30 mg B.D, days 1-21 every 28 days, with crizotinib 250 mg O.D continuously. Dose-limiting toxicities included grade ≥ 3 transaminitis, creatinine phosphokinase increases and fatigue. Thirty-six RASMT metastatic CRC patients were enrolled in the dose expansion. Pharmacokinetic and pharmacodynamic parameters showed evidence of target engagement. Across the entire study, the most frequent treatment-related adverse events (TR-AE) were rash (80.4%), fatigue (53.4%) and diarrhoea (51.8%) with grade ≥ 3 TR-AE occurring in 44.6%. Best clinical response within the RASMT CRC cohort was stable disease in seven patients (24%). Tumour MET super-expression (IHC H-score > 180 and MET ISH + 3) was observed in 7 patients (24.1%), with MET-amplification only present in 1 of these patients. This patient discontinued treatment early during cycle 1 due to toxicity. Patients with high baseline RASMT allele frequency had a significant shorter median overall survival compared with that seen for patients with low baseline KRASMT allele frequency. CONCLUSIONS Combination binimetinib/crizotinib showed a poor tolerability with no objective responses observed in RASMT advanced CRC patients. EudraCT-Number: 2014-000463 - 40 (20/06/2014: A Sequential Phase I study of MEK1/2 inhibitors PD- 0325901 or Binimetinib combined with cMET inhibitor Crizotinib in RAS Mutant and RAS Wild Type with aberrant c-MET).
Collapse
Affiliation(s)
- Francesca Aroldi
- Department of Oncology, University of Oxford, Old Road Campus Research Building Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Elena Elez
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Thierry André
- Department of Medical Oncology, Sorbonne Université, Hôpital Saint Antoine, 75012, Paris, France
| | - Geraldine Perkins
- Department of GI Oncology, Hôpital Européen Georges-Pompidou, 75015, Paris, France
| | - Hans Prenen
- Department of Medical Oncology, University of Antwerp/Antwerp University Hospital, 2610, Wilrijk, Belgium
| | - Vlad Popovici
- Faculty of Science, RECETOX, Masaryk University, 625 00, Brno, Czech Republic
| | - Peter Gallagher
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Jennifer Houlden
- Oncology Clinical Trials Office (OCTO), Department of Oncology, University of Oxford, Oxford, OX3 7LJ, UK
| | - Linda Collins
- Oncology Clinical Trials Office (OCTO), Department of Oncology, University of Oxford, Oxford, OX3 7LJ, UK
| | - Corran Roberts
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Christian Rolfo
- Department of Medical Oncology, University of Antwerp/Antwerp University Hospital, 2610, Wilrijk, Belgium
| | - Federica Di Nicolantonio
- Department of Oncology &, University of Torino, Candiolo Cancer Institute, 10060, Candiolo, TO, Italy
| | - Margaret Grayson
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK
| | - Ruth Boyd
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK
| | - Karolien Bettens
- Genomics, Diagnostics and Genomics Group, Agilent Technologies, 1831, Diegem, Belgium
| | - Jurgen Delfavero
- Genomics, Diagnostics and Genomics Group, Agilent Technologies, 1831, Diegem, Belgium
| | - Victoria Coyle
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Mark Lawler
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Hajrah Khawaja
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Pierre Laurent-Puig
- Institut National de La Sante Et de La Recherche Medicale (INSERM), Universite Paris Descartes, 75006, Paris, France
| | - Manuel Salto-Tellez
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Tim S Maughan
- Department of Oncology, University of Oxford, Old Road Campus Research Building Roosevelt Drive, Oxford, OX3 7DQ, UK
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton St, Liverpool, L69 3GE, UK
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Richard Adams
- Cardiff University and Velindre University NHS Trust, Cardiff, CF14 2 TL, UK
| | - Robert Jones
- Cardiff University and Velindre University NHS Trust, Cardiff, CF14 2 TL, UK
| | - Bryan T Hennessy
- Royal College of Surgeons in Ireland University of Medicine and Health Sciences, 123 St. Stephen's, Green, Dublin, Ireland
| | - Alberto Bardelli
- Department of Oncology, Molecular Biotechnology Center, University of Torino, Turin, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marc Peeters
- Department of Medical Oncology, University of Antwerp/Antwerp University Hospital, 2610, Wilrijk, Belgium
| | - Mark R Middleton
- Department of Oncology, University of Oxford, Old Road Campus Research Building Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Richard H Wilson
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Sandra Van Schaeybroeck
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, BT9 7 AB, UK.
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, BT9 7AE, UK.
| |
Collapse
|
6
|
Hazariwala V, Rossen JL, Lasky-Zeid J, Waanders A, Bohnsack BL. Baseline ophthalmic findings and complications in pediatric patients treated with MEK inhibitors. J AAPOS 2025; 29:104157. [PMID: 40032239 DOI: 10.1016/j.jaapos.2025.104157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/21/2024] [Accepted: 12/15/2024] [Indexed: 03/05/2025]
Abstract
BACKGROUND MEK inhibitors are an increasingly used class of medications associated with ocular complications, including serous retinal detachments, vein occlusions, uveitis, and dry eye syndrome in adults. However, baseline ophthalmic findings and MEK inhibitor-related complications in children are underreported. METHODS The medical records of patients <21 years of age who underwent MEK inhibitor treatment at our institution between 2016 and 2023 were reviewed retrospectively. Patients with baseline examination and at least one follow-up eye examination were included. Systemic and ophthalmic diagnoses, MEK inhibitor treatment course, and ocular findings were extracted from the record. RESULTS A total of 45 patients (23 males) were treated with a MEK inhibitor for tumor with neurofibromatosis type 1 (NF1, n = 20), tumor without NF1 (n = 22), or non-tumor (n = 3) at a mean age of 9.4 ± 4.8 years. Nineteen patients were treated with trametinib, 20 with selumetinib, and 6 with combined trametinib and dabrafenib. Thirty-three patients (73%), all with neurological tumors, had abnormal baseline eye examinations, including optic nerve abnormalities (n = 20), visual acuity impairment (n = 17), visual field defect (n = 8), strabismus (n = 10), or anterior segment abnormalities (n = 14). Average time on MEK inhibitors was 445.5 ± 377.8 days. Three patients with neurologic tumors without NF1 developed ophthalmic side effects of dry eye syndrome (n = 1) or symptomatic anterior uveitis (n = 2) at 6-7 months after initiation of MEK inhibitor. All 3 patients responded to topical medications. CONCLUSIONS The majority of children started on MEK inhibitors had baseline eye abnormalities due to ocular or visual pathway involvement of neurological tumors. Ophthalmic side effects of MEK inhibitors occurred in 7% of children. Additional studies are required to formulate screening guidelines for the pediatric population.
Collapse
Affiliation(s)
- Vikisha Hazariwala
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jennifer L Rossen
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Janice Lasky-Zeid
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Angela Waanders
- Division of Hematology, Oncology, NeuroOncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Brenda L Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois; Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
7
|
Alotaibi HA, Yuen HK, Alshaibani AK, Alibrahim EA. Selumetinib use as targeted therapy for plexiform neurofibroma: a comprehensive review of the literature. Orbit 2025:1-8. [PMID: 40019319 DOI: 10.1080/01676830.2025.2470989] [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: 11/18/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
Neurofibromatosis type 1 is inherited in an autosomal dominant fashion. The approximate prevalence of neurofibromatosis type 1 is 1:2500 to 1:3500 regardless of ethnic group. Around 50% of neurofibromatosis type 1 patients develop plexiform neurofibroma, a lesion that has a tendency for malignant transformation. Understanding the pathophysiology of plexiform neurofibromatosis and the implication of neurofibromin signal transduction has led to the development and use of targeted therapy on this pathway. In this article, we comprehensively reviewed the recently published literature on the use of selumetinib to treat inoperable plexiform neurofibroma in neurofibromatosis type 1 patients.
Collapse
Affiliation(s)
- Hammam A Alotaibi
- Department of Ophthalmology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hunter Kl Yuen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Askar K Alshaibani
- Department of Ophthalmology, Dhahran Eye Specialist Hospital, Dhahran, Saudi Arabia
| | - Eman A Alibrahim
- Department of Ophthalmology, Dhahran Eye Specialist Hospital, Dhahran, Saudi Arabia
| |
Collapse
|
8
|
Baruah V, Krishna BA, Kelly MC, Qi X, O’Connor CM. Inhibition of MAPK signaling suppresses cytomegalovirus reactivation in CD34 + Kasumi-3 cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638080. [PMID: 39990446 PMCID: PMC11844548 DOI: 10.1101/2025.02.13.638080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Reactivation of latent human cytomegalovirus (CMV) can lead to severe complications in individuals with dysregulated immune systems. While antiviral therapies for CMV are approved, these compounds are limited by their toxicity and inability to specifically target the latent reservoir or prevent reactivation. Herein we show that CMV reactivation in Kasumi-3 cells, a CD34+ hematopoietic cell line, requires mitogen-activated protein kinase (MAPK) activation. Importantly, pharmacological inhibition of the MAPK signaling pathway, including MEK and ERK, restricts viral reactivation in Kasumi-3 cells. In sum, our findings show MAPK signaling is critical for CMV reactivation, revealing a potential avenue for therapeutic intervention to prevent viral reactivation and downstream pathogenesis that is often detrimental for immunosuppressed and immunocompromised patients.
Collapse
Affiliation(s)
- Vargab Baruah
- Infection Biology; Sheikha Fatima bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195 USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106 USA
| | - Benjamin A. Krishna
- Infection Biology; Sheikha Fatima bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195 USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106 USA
| | - Michael C. Kelly
- Infection Biology; Sheikha Fatima bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195 USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106 USA
| | - Xu Qi
- Infection Biology; Sheikha Fatima bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195 USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106 USA
| | - Christine M. O’Connor
- Infection Biology; Sheikha Fatima bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195 USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106 USA
- Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH 44195 USA
| |
Collapse
|
9
|
Yang Y, Yang H, Gao Y, Yang Q, Zhu X, Miao Q, Xu X, Li Z, Zuo D. EML4-ALK G1202R and EML4-ALK L1196M mutations induce crizotinib resistance in non-small cell lung cancer cells through activating epithelial-mesenchymal transition mediated by MDM2/MEK/ERK signal axis. Cell Biol Int 2025; 49:55-67. [PMID: 39318039 DOI: 10.1002/cbin.12249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 08/04/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Crizotinib, as the first-generation of anaplastic lymphoma kinase (ALK) inhibitor, effectively improves the survival time of ALK-positive non-small cell lung cancer (NSCLC) patients. However, its efficacy is severely limited by drug resistance caused by secondary mutations. G1202R and L1196M are classical mutation sites located in ALK kinase domain. They may hinder the binding of ALK inhibitors to the target kinase domain, resulting in drug resistance in patients. However, the exact mechanism of drug resistance mediated by these mutations remains unclear. In this study, we aimed to evaluate how G1202R and L1196M mutations mediate crizotinib resistance. To explore the resistance mechanism, we constructed EML4-ALK G1202R and L1196M mutant cell lines with A549 cells. The results showed that the mutant cells exhibited significant epithelial-mesenchymal transition (EMT) and metastasis compared to control (A549-vector) or wild type (A549-EML4-ALK) cells. Subsequently, it was found that the occurrence of EMT was correlated to the high expression of murine double minute 2 (MDM2) protein and the activation of mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway in mutant cells. Down-regulation of MDM2 inhibited the activation of MEK/ERK pathway, thus reversed the EMT process and markedly increased the inhibitory effect of crizotinib on the growth of mutant cells. Collectively, resistance of ALK-positive NSCLC cells to crizotinib is induced by G1202R and L1196M mutations through activation of the MDM2/MEK/ERK signalling axis, promoting EMT process and metastasis. These findings suggest that the combination of MDM2 inhibitors and crizotinib could be a potential therapeutic strategy.
Collapse
MESH Headings
- Crizotinib/pharmacology
- Humans
- Epithelial-Mesenchymal Transition/drug effects
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Proto-Oncogene Proteins c-mdm2/metabolism
- Proto-Oncogene Proteins c-mdm2/genetics
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/genetics
- MAP Kinase Signaling System/drug effects
- Mutation
- Animals
- A549 Cells
- Cell Line, Tumor
- Mice, Nude
- Mice
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Yuying Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Huan Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yunhui Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Qian Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinya Zhu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Qianying Miao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaobo Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
10
|
Rekowski J, Guo C, Solovyeva O, Dimairo M, Rouhifard M, Patel D, Alger E, Ashby D, Berlin J, Boix O, Calvert M, Chan AW, Coschi CH, de Bono J, Evans TRJ, Garrett–Mayer E, Golub RM, Hayward KS, Hopewell S, Isaacs JD, Ivy SP, Jaki T, Kholmanskikh O, Kightley A, Lee S, Liu R, Maia I, Mander A, Marshall LV, Matcham J, Peck R, Rantell KR, Richards DP, Seymour L, Tanaka Y, Ursino M, Weir CJ, Yap C. CONSORT-DEFINE explanation and elaboration: recommendations for enhancing reporting quality and impact of early phase dose-finding clinical trials. EClinicalMedicine 2025; 79:102987. [PMID: 39877553 PMCID: PMC11773258 DOI: 10.1016/j.eclinm.2024.102987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/31/2025] Open
Abstract
Early phase dose-finding (EPDF) trials are key in the development of novel therapies, with their findings directly informing subsequent clinical development phases and providing valuable insights for reverse translation. Comprehensive and transparent reporting of these studies is critical for their accurate and critical interpretation, which may improve and expedite therapeutic development. However, quality of reporting of design characteristics and results from EPDF trials is often variable and incomplete. The international consensus-based CONSORT-DEFINE (Consolidated Standards for Reporting Trials Dose-finding Extension) statement, an extension of the CONSORT statement for randomised trials, was developed to improve the reporting of EPDF trials. The CONSORT-DEFINE statement introduced 21 new items and modified 19 existing CONSORT items.This CONSORT-DEFINE Explanation and Elaboration (E&E) document provides important information to enhance understanding and facilitate the implementation of the CONSORT-DEFINE checklist. For each new or modified checklist item, we provide a detailed description and its rationale with supporting evidence, and present examples from EPDF trial reports published in peer-reviewed scientific journals. When reporting the results of EPDF trials, authors are encouraged to consult the CONSORT-DEFINE E&E document, together with the CONSORT and CONSORT-DEFINE statement papers, and adhere to their recommendations. Widespread adoption of the CONSORT-DEFINE statement is likely to enhance the reporting quality of EPDF trials, thus facilitating the peer review of such studies and their appraisal by researchers, regulators, ethics committee members, and funders. Funding This work is a further extension of the CONSORT-DEFINE study, which was funded by the UK Medical Research Council (MRC)-National Institute for Health and Care Research (NIHR) Methodology Research Programme (MR/T044934/1). The Clinical Trials and Statistics Unit at The Institute of Cancer Research (ICR-CTSU) receives programmatic infrastructure funding from Cancer Research UK (C1491/A25351; CTUQQR-Dec 22/100 004), which has contributed to accelerating the advancement and successful completion of this work.
Collapse
Affiliation(s)
- Jan Rekowski
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Christina Guo
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Olga Solovyeva
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Munyaradzi Dimairo
- Division of Population Health, Sheffield Centre for Health and Related Research, University of Sheffield, Sheffield, UK
| | - Mahtab Rouhifard
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Dhrusti Patel
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Emily Alger
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| | - Deborah Ashby
- School of Public Health, Imperial College London, St Mary's Hospital, London, UK
| | | | | | - Melanie Calvert
- Centre for Patient Reported Outcomes Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Applied Research Collaboration West Midlands, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Precision Transplant and Cellular Therapeutics, University of Birmingham, Birmingham, UK
- National Institute for Health and Care Research Birmingham Biomedical Research Centre, NIHR Birmingham Biomedical Research Centre, Institute of Translational Medicine, University Hospital NHS Foundation Trust, Birmingham, UK
| | - An-Wen Chan
- Department of Medicine, Women's College Research Institute, University of Toronto, Toronto, Canada
| | | | - Johann de Bono
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Thomas R. Jeffry Evans
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Elizabeth Garrett–Mayer
- Center for Research and Analytics, American Society of Clinical Oncology, Alexandria, VA, USA
| | - Robert M. Golub
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn S. Hayward
- Departments of Physiotherapy and Medicine, University of Melbourne, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Sally Hopewell
- Oxford Clinical Research Unit, NDORMS, University of Oxford, Oxford, UK
| | - John D. Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle upon Tyne, UK
| | - S. Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Institute of Health, Bethesda, MD, USA
| | - Thomas Jaki
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK
- Computational Statistics Group, University of Regensburg, Regensburg, Germany
| | | | - Andrew Kightley
- Patient and Public Involvement and Engagement (PPIE) Lead, Lichfield, UK
| | - Shing Lee
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | | | - Adrian Mander
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Lynley V. Marshall
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - James Matcham
- Strategic Consulting, Cytel (Australia), Perth, WA, Australia
| | - Richard Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Hoffmann-La Roche, Basel, Switzerland
| | | | | | | | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Moreno Ursino
- ReCAP/F CRIN, INSERM, 5400, Nancy, France
- Unit of Clinical Epidemiology, University Hospital Centre Robert Debré, Université Paris Cité, Paris, France
- INSERM, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- HeKA Team, Centre Inria, Paris, France
| | - Christopher J. Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Christina Yap
- Clinical Trials and Statistics Unit at the Institute of Cancer Research, London, UK
| |
Collapse
|
11
|
Lixi F, Giannaccare G, Salerno G, Gagliardi V, Pellegrino A, Vitiello L. Side Effects of Novel Anticancer Drugs on the Posterior Segment of the Eye: A Review of the Literature. J Pers Med 2024; 14:1160. [PMID: 39728071 DOI: 10.3390/jpm14121160] [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: 11/21/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Currently, common treatment approaches for neoplastic diseases include surgery, radiation, and/or anticancer drugs (chemotherapy, hormone medications, and targeted therapies). In particular, anticancer medicines destroy cancerous cells by blocking certain pathways that aid in the disease's initiation and progression. These pharmaceutical drugs' capacity to inhibit malignant cells has made them indispensable in the treatment of neoplastic disorders. Nonetheless, considering their cyto- and neurotoxicity, as well as their inflammatory responses, these medications may also have unfavorable systemic and ocular side effects. In fact, it is well known that ocular posterior segment side effects, including retinal and vascular complications, have a negative influence on the patient's eyesight and quality of life. However, the underlying mechanisms contributing to the development of these side effects remain incompletely recognized, especially in the case of newly available anticancer drugs. The purpose of this literature review is to analyze the possible side effects of new anticancer drugs on the posterior segment of the eye, trying to better understand the involved pharmacological mechanisms and offer helpful guidance on their appropriate management.
Collapse
Affiliation(s)
- Filippo Lixi
- Eye Clinic, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, CA, Italy
| | - Giuseppe Giannaccare
- Eye Clinic, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, CA, Italy
| | - Giulio Salerno
- Eye Unit, "Luigi Curto" Hospital, Azienda Sanitaria Locale Salerno, 84035 Polla, SA, Italy
| | - Vincenzo Gagliardi
- Eye Unit, "Luigi Curto" Hospital, Azienda Sanitaria Locale Salerno, 84035 Polla, SA, Italy
| | - Alfonso Pellegrino
- Eye Unit, "Luigi Curto" Hospital, Azienda Sanitaria Locale Salerno, 84035 Polla, SA, Italy
| | - Livio Vitiello
- Eye Unit, "Luigi Curto" Hospital, Azienda Sanitaria Locale Salerno, 84035 Polla, SA, Italy
| |
Collapse
|
12
|
Koshrovski-Michael S, Ajamil DR, Dey P, Kleiner R, Tevet S, Epshtein Y, Green Buzhor M, Khoury R, Pozzi S, Shenbach-Koltin G, Yeini E, Woythe L, Blau R, Scomparin A, Barshack I, Florindo HF, Lazar S, Albertazzi L, Amir RJ, Satchi-Fainaro R. Two-in-one nanoparticle platform induces a strong therapeutic effect of targeted therapies in P-selectin-expressing cancers. SCIENCE ADVANCES 2024; 10:eadr4762. [PMID: 39671487 PMCID: PMC11641104 DOI: 10.1126/sciadv.adr4762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Combined therapies in cancer treatment aim to enhance antitumor activity. However, delivering multiple small molecules imposes challenges, as different drugs have distinct pharmacokinetic profiles and tumor penetration abilities, affecting their therapeutic efficacy. To circumvent this, poly(lactic-co-glycolic acid) (PLGA)-polyethylene glycol (PEG)-based nanoparticles were developed as a platform for the codelivery of synergistic drug ratios, improving therapeutic efficacy by increasing the percentage of injected dose reaching the tumor. Nonetheless, extravasation-dependent tumor accumulation is susceptible to variations in tumor vasculature; therefore, PLGA-PEG was modified with sulfates to actively target P-selectin-expressing cancers. Here, we show the potential of our platform in unique three-dimensional (3D) in vitro and in vivo models. The P-selectin-targeted nanoparticles showed enhanced accumulation in 3D spheroids and tissues of P-selectin-expressing BRAF-mutated melanomas and BRCA-mutated breast cancers, resulting in superior in vivo efficacy and safety. This nanoplatform could advance the codelivery of a plethora of anticancer drug combinations to various P-selectin-expressing tumors.
Collapse
Affiliation(s)
- Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Chemistry, Siksha Bhavana, Visva-Bharati University, Santiniketan, West Bengal 731235, India
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shahar Tevet
- The Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- ADAMA Center for Novel Delivery Systems in Crop Protection, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yana Epshtein
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Marina Green Buzhor
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gal Shenbach-Koltin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Laura Woythe
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology (TU/e), 5612AZ Eindhoven, Netherlands
| | - Rachel Blau
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Turin 10125, Italy
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-003, Portugal
| | - Shlomi Lazar
- Department of Pharmacology, Israel Institute of Biological Research (IIBR), Ness-Ziona 74100, Israel
| | - Lorenzo Albertazzi
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology (TU/e), 5612AZ Eindhoven, Netherlands
| | - Roey J. Amir
- The Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- ADAMA Center for Novel Delivery Systems in Crop Protection, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
13
|
Ghosh S, Fan F, Powell R, Park YS, Stephan C, Kopetz ES, Ellis LM, Bhattacharya R. Enhancing efficacy of the MEK inhibitor trametinib with paclitaxel in KRAS-mutated colorectal cancer. Ther Adv Med Oncol 2024; 16:17588359241303302. [PMID: 39664300 PMCID: PMC11632859 DOI: 10.1177/17588359241303302] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Background KRAS is frequently mutated in the tumors of patients with metastatic colorectal cancer (mCRC) and thus represents a valid target for therapy. However, the strategies of targeting KRAS directly and targeting the downstream effector mitogen-activated protein kinase kinase (MEK) via monotherapies have shown limited efficacy. Thus, there is a strong need for novel, effective combination therapies to improve MEK-inhibitor efficacy in patients with KRAS-mutated mCRC. Objective Our objective was to identify novel drug combinations that enhance MEK-inhibitor efficacy in patients with KRAS-mutated mCRC. Design In this study, we performed unbiased high-throughput screening (HTS) to identify drugs that enhance the efficacy of MEK inhibitors in vitro, and we validated the drugs' efficacy in vivo. Methods HTS was performed using three-dimensional CRC spheroids. Trametinib, the anchor drug, was probed with two "clinically ready" libraries of 252 drugs to identify effective drug combinations. The effects of the drug combinations on CRC cell proliferation and apoptosis were further validated using cell growth assays, flow cytometry, and biochemical assays. Proteomic and immunostaining studies were performed to determine the drugs' effects on molecular signaling and cell division. The effects of the drug combinations were examined in vivo using CRC patient-derived xenografts. Results HTS identified paclitaxel as being synergistic with trametinib. In vitro validation showed that, compared with monotherapies, this drug combination demonstrated strong inhibition of cell growth, reduced colony formation, and enhanced apoptosis in multiple KRAS-mutated CRC cell lines. Mechanistically, combining trametinib with paclitaxel led to alterations in signaling mediators that block cell-cycle progression. Trametinib also enhanced paclitaxel-mediated microtubule stability resulting in significantly higher defects in mitosis. Finally, the combination of trametinib with paclitaxel exhibited significant inhibition of tumor growth in several KRAS-mutant patient-derived xenograft mouse models. Conclusion Our data provide evidence supporting clinical trials of trametinib with paclitaxel as a novel therapeutic option for patients with KRAS-mutated, metastatic CRC.
Collapse
Affiliation(s)
- Susmita Ghosh
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fan Fan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Reid Powell
- Department of Translational Medical Sciences, Texas A&M University School of Medicine, Houston, TX, USA
| | - Yong Sung Park
- Department of Translational Medical Sciences, Texas A&M University School of Medicine, Houston, TX, USA
| | - Clifford Stephan
- Department of Translational Medical Sciences, Texas A&M University School of Medicine, Houston, TX, USA
| | - E. Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lee M. Ellis
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rajat Bhattacharya
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| |
Collapse
|
14
|
Pascarella A, Limongelli G, De Falco A, Minale EMP, Di Nardo G, Di Marco GM, Zito Marinosci G, Olimpico G, Siani P, De Brasi D. Refractory Chylothorax and Ventricular Hypertrophy Treated with Trametinib in a Patient with Noonan Syndrome: 18-Month Follow-Up. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1342. [PMID: 39594917 PMCID: PMC11593188 DOI: 10.3390/children11111342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024]
Abstract
RASopathies are a group of genetic syndromes caused by germline mutations in genes involved in the RAS/Mitogen-Activated Protein Kinase signaling pathway, which regulates cellular proliferation, differentiation, and angiogenesis. Despite their involvement at different levels of this pathway, RASopathies share overlapping clinical phenotypes. Noonan syndrome is the most prevalent RASopathy, with an estimated incidence of 1 in 2500 live births, and it is typically inherited in an autosomal dominant manner, with 50% of cases involving gain-of-function mutations in the PTPN11 gene. De novo mutations are common, accounting for 60% of cases. The phenotype of Noonan syndrome includes characteristic facial and physical features, congenital cardiac defects, lymphatic and cerebrovascular anomalies, renal malformations, hematological abnormalities, developmental issues, and an increased risk of cancer. Severe congenital cardiac defects and lymphatic abnormalities significantly impact prognosis, contributing to increased morbidity and mortality. Recent therapeutic advancements have introduced trametinib, an MEK1/2 inhibitor, for treating Noonan syndrome patients with severe cardiac and lymphatic complications. To assess its efficacy, here, we present a case of a newborn with Noonan syndrome who exhibited refractory chylothorax, ventricular hypertrophy, and pulmonary stenosis who was treated with trametinib. The patient demonstrated significant improvement in chylothorax and left ventricular hypertrophy, though pulmonary stenosis persisted. This case further confirms trametinib's potential as a therapeutic option for severe Noonan syndrome complications, emphasizing the need for further clinical trials to optimize treatment protocols and evaluate long-term outcomes.
Collapse
Affiliation(s)
- Antonia Pascarella
- Unit of Chronic and Multifactorial Diseases, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (A.P.); (P.S.)
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Diseases Unit, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 81031 Naples, Italy
| | | | | | - Giangiacomo Di Nardo
- Department of Pediatric Cardiology, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (G.D.N.)
| | - Giovanni Maria Di Marco
- Department of Pediatric Cardiology, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (G.D.N.)
| | | | - Giorgia Olimpico
- Department of Translational Medical Science, Section of Pediatrics, University “Federico II”, 80131 Naples, Italy;
| | - Paolo Siani
- Unit of Chronic and Multifactorial Diseases, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (A.P.); (P.S.)
| | - Daniele De Brasi
- Medical Genetics Unit, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy;
| |
Collapse
|
15
|
Ben-Yishay R, Globus O, Balint-Lahat N, Arbili-Yarhi S, Bar-Hai N, Bar V, Aharon S, Kosenko A, Zundelevich A, Berger R, Ishay-Ronen D. Class Effect Unveiled: PPARγ Agonists and MEK Inhibitors in Cancer Cell Differentiation. Cells 2024; 13:1506. [PMID: 39273076 PMCID: PMC11394433 DOI: 10.3390/cells13171506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) plays a major role in breast cancer progression and the development of drug resistance. We have previously demonstrated a trans-differentiation therapeutic approach targeting invasive dedifferentiated cancer cells. Using a combination of PPARγ agonists and MEK inhibitors, we forced the differentiation of disseminating breast cancer cells into post-mitotic adipocytes. Utilizing murine breast cancer cells, we demonstrated a broad class effect of PPARγ agonists and MEK inhibitors in inducing cancer cell trans-differentiation into adipocytes. Both Rosiglitazone and Pioglitazone effectively induced adipogenesis in cancer cells, marked by PPARγ and C/EBPα upregulation, cytoskeleton rearrangement, and lipid droplet accumulation. All tested MEK inhibitors promoted adipogenesis in the presence of TGFβ, with Cobimetinib showing the most prominent effects. A metastasis ex vivo culture from a patient diagnosed with triple-negative breast cancer demonstrated a synergistic upregulation of PPARγ with the combination of Pioglitazone and Cobimetinib. Our results highlight the potential for new therapeutic strategies targeting cancer cell plasticity and the dedifferentiation phenotype in aggressive breast cancer subtypes. Combining differentiation treatments with standard therapeutic approaches may offer a strategy to overcome drug resistance.
Collapse
Affiliation(s)
- Rakefet Ben-Yishay
- Oncology Institute, Sheba Medical Center, Ramat Gan 5262000, Israel; (R.B.-Y.); (O.G.); (S.A.-Y.); (N.B.-H.)
| | - Opher Globus
- Oncology Institute, Sheba Medical Center, Ramat Gan 5262000, Israel; (R.B.-Y.); (O.G.); (S.A.-Y.); (N.B.-H.)
| | - Nora Balint-Lahat
- Institute of Pathology, Sheba Medical Center, Ramat Gan 5262000, Israel;
| | - Sheli Arbili-Yarhi
- Oncology Institute, Sheba Medical Center, Ramat Gan 5262000, Israel; (R.B.-Y.); (O.G.); (S.A.-Y.); (N.B.-H.)
| | - Neta Bar-Hai
- Oncology Institute, Sheba Medical Center, Ramat Gan 5262000, Israel; (R.B.-Y.); (O.G.); (S.A.-Y.); (N.B.-H.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Vered Bar
- Curesponse Ltd., Rehovot 7670102, Israel; (V.B.); (A.K.)
| | - Sara Aharon
- Curesponse Ltd., Rehovot 7670102, Israel; (V.B.); (A.K.)
| | - Anna Kosenko
- Curesponse Ltd., Rehovot 7670102, Israel; (V.B.); (A.K.)
| | | | - Raanan Berger
- Oncology Institute, Sheba Medical Center, Ramat Gan 5262000, Israel; (R.B.-Y.); (O.G.); (S.A.-Y.); (N.B.-H.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Ishay-Ronen
- Oncology Institute, Sheba Medical Center, Ramat Gan 5262000, Israel; (R.B.-Y.); (O.G.); (S.A.-Y.); (N.B.-H.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
16
|
Ghosh S, Fan F, Powell R, Park Y, Stephan C, Kopetz ES, Ellis LM, Bhattacharya R. Enhancing efficacy of the MEK inhibitor trametinib with paclitaxel in KRAS -mutated colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611458. [PMID: 39282351 PMCID: PMC11398522 DOI: 10.1101/2024.09.05.611458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Background KRAS is frequently mutated in the tumors of patients with metastatic colorectal cancer (mCRC) and thus represents a valid target for therapy. However, the strategies of targeting KRAS directly and targeting the downstream effector mitogen-activated protein kinase kinase (MEK) via monotherapies have shown limited efficacy. Thus, there is a strong need for novel, effective combination therapies to improve MEK-inhibitor efficacy in patients with KRAS -mutated mCRC. Objective Our objective was to identify novel drug combinations that enhance MEK-inhibitor efficacy in patients with KRAS -mutated mCRC. Design In this study, we performed unbiased high-throughput screening (HTS) to identify drugs that enhance the efficacy of MEK inhibitors in vitro , and we validated the efficacy of the drugs in vivo . Methods HTS was performed using 3-dimensional CRC spheroids. Trametinib, the anchor drug, was probed with 2 clinically ready libraries of 252 drugs to identify effective drug combinations. The effects of the drug combinations on CRC cell proliferation and apoptosis were further validated using cell growth assays, flow cytometry, and biochemical assays. Proteomic and immunostaining studies were performed to determine the effects of the drugs on molecular signaling and cell division. The effects of the drug combinations were examined in vivo using CRC patient-derived xenografts. Results HTS identified paclitaxel as being synergistic with trametinib. In vitro validation showed that, compared with monotherapies, this drug combination demonstrated strong inhibition of cell growth, reduced colony formation, and enhanced apoptosis in multiple KRAS -mutated CRC cell lines. Mechanistically, combining trametinib with paclitaxel led to alterations in signaling mediators that block cell cycle progression and increases in microtubule stability that resulted in significantly higher defects in the mitosis. Finally, the combination of trametinib with paclitaxel exhibited significant inhibition of tumor growth in several KRAS -mutant patient-derived xenograft mouse models. Conclusion Our data provide evidence supporting clinical trials of trametinib with paclitaxel as a novel therapeutic option for patients with KRAS -mutated, metastatic CRC.
Collapse
|
17
|
Karasawa H, Yasumizu Y, Kosaka T, Shimoi T, Oya M. Efficacy of trametinib in a metastatic urothelial carcinoma patient with a BRAF mutation. IJU Case Rep 2024; 7:375-378. [PMID: 39224677 PMCID: PMC11366432 DOI: 10.1002/iju5.12759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction BRAF mutations in bladder cancer are rare. MEK inhibitors have excellent clinical benefits in the treatment of melanoma. Case presentation A 60-year-old male was diagnosed with muscle-invasive bladder cancer and underwent total cystectomy and ileal conduit diversion. Despite 4 cycles of gemcitabine and cisplatin chemotherapy and 3 courses of pembrolizumab, the left obturator lymph node enlarged. Cancer multi-gene panel testing confirmed the BRAF G469A mutation and trametinib was recommended. Three months after the initiation of trametinib (2 mg, qd), the left obturator lymph node shrank by more than 50%. The disease has remained stable for more than 18 months. Conclusion The present case indicates the potential of trametinib to treat mBUC patients with the BRAF G469A mutation in this setting.
Collapse
Affiliation(s)
| | - Yota Yasumizu
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Takeo Kosaka
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Tatsunori Shimoi
- Department of Breast and Medical OncologyNational Cancer HospitalTokyoJapan
| | - Mototsugu Oya
- Department of UrologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
18
|
Decaudin D, Némati F, Masliah Planchon J, Seguin-Givelet A, Lefevre M, Etienne V, Ahnine H, Peretti Q, Sourd L, El-Botty R, Huguet L, Lagha S, Hegarat N, Roman-Roman S, Bièche I, Girard N, Montaudon E. Evaluation of Combined Chemotherapy and Genomic-Driven Targeted Therapy in Patient-Derived Xenografts Identifies New Therapeutic Approaches in Squamous Non-Small-Cell Lung Cancer Patients. Cancers (Basel) 2024; 16:2785. [PMID: 39199558 PMCID: PMC11352497 DOI: 10.3390/cancers16162785] [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: 07/09/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of chemotherapy and targeted therapy has been validated in non-small-cell lung cancer (NSCLC) patients with EGFR mutations. We therefore investigated whether this type of combined approach could be more widely used by targeting other genetic alterations present in NSCLC. PDXs were generated from patients with NSCLC adenocarcinomas (ADCs) and squamous-cell carcinomas (SCCs). Targeted NGS analyses identified various molecular abnormalities in the MAPK and PI3K pathways and in the cell cycle process in our PDX panel. The antitumor efficacy of targeted therapies alone or in combination with chemotherapy was then tested in vivo. We observed that trametinib, BKM120, AZD2014 and palbociclib increased the efficacy of each chemotherapy in SCC PDXs, in contrast to a non-insignificant or slight improvement in ADCs. Furthermore, we observed high efficacy of trametinib in KRAS-, HRAS- and NRAS-mutated tumors (ADCs and SCCs), suggesting that the MEK inhibitor may be useful in a wider population of NSCLC patients, not just those with KRAS-mutated ADCs. Our results suggest that the detection of pathogenic variants by NGS should be performed in all NSCLCs, and particularly in SCCs, to offer patients a more effective combination of chemotherapy and targeted therapy.
Collapse
Affiliation(s)
- Didier Decaudin
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
| | - Fariba Némati
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | | | - Agathe Seguin-Givelet
- Department of Thoracic Surgery, Curie-Montsouris Thorax Institute, Institut Mutualiste Montsouris, 75014 Paris, France;
- Faculty of Medicine SMBH, Paris 13 University, Sorbonne Paris Cité, 75013 Bobigny, France
| | - Marine Lefevre
- Department of Pathology, Institut Mutualiste Montsouris, 75014 Paris, France;
| | - Vesnie Etienne
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Harry Ahnine
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Quentin Peretti
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Laura Sourd
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Rania El-Botty
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Lea Huguet
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| | - Sarah Lagha
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
| | - Nadia Hegarat
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
| | - Sergio Roman-Roman
- Department of Translationnal Research, Institut Curie, PSL University Paris, 75006 Paris, France;
| | - Ivan Bièche
- Department of Genetic, Institut Curie, 75005 Paris, France; (J.M.P.); (I.B.)
| | - Nicolas Girard
- Department of Medical Oncology, Institut Curie, 75005 Paris, France; (S.L.); (N.H.); (N.G.)
- Paris Saclay University, University of Versailles Saint-Quentin-en-Yvelines (UVSQ), 91405 Versailles, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie, PSL University Paris, 75005 Paris, France; (F.N.); (V.E.); (L.S.); (R.E.-B.); (L.H.)
| |
Collapse
|
19
|
Kotani H, Oshima H, Boucher JC, Yamano T, Sakaguchi H, Sato S, Fukuda K, Nishiyama A, Yamashita K, Ohtsubo K, Takeuchi S, Nishiuchi T, Oshima M, Davila ML, Yano S. Dual inhibition of SUMOylation and MEK conquers MYC-expressing KRAS-mutant cancers by accumulating DNA damage. J Biomed Sci 2024; 31:68. [PMID: 38992694 PMCID: PMC11238369 DOI: 10.1186/s12929-024-01060-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND KRAS mutations frequently occur in cancers, particularly pancreatic ductal adenocarcinoma, colorectal cancer, and non-small cell lung cancer. Although KRASG12C inhibitors have recently been approved, effective precision therapies have not yet been established for all KRAS-mutant cancers. Many treatments for KRAS-mutant cancers, including epigenome-targeted drugs, are currently under investigation. Small ubiquitin-like modifier (SUMO) proteins are a family of small proteins covalently attached to and detached from other proteins in cells via the processes called SUMOylation and de-SUMOylation. We assessed whether SUMOylation inhibition was effective in KRAS-mutant cancer cells. METHODS The efficacy of the first-in-class SUMO-activating enzyme E inhibitor TAK-981 (subasumstat) was assessed in multiple human and mouse KRAS-mutated cancer cell lines. A gene expression assay using a TaqMan array was used to identify biomarkers of TAK-981 efficacy. The biological roles of SUMOylation inhibition and subsequent regulatory mechanisms were investigated using immunoblot analysis, immunofluorescence assays, and mouse models. RESULTS We discovered that TAK-981 downregulated the expression of the currently undruggable MYC and effectively suppressed the growth of MYC-expressing KRAS-mutant cancers across different tissue types. Moreover, TAK-981-resistant cells were sensitized to SUMOylation inhibition via MYC-overexpression. TAK-981 induced proteasomal degradation of MYC by altering the balance between SUMOylation and ubiquitination and promoting the binding of MYC and Fbxw7, a key factor in the ubiquitin-proteasome system. The efficacy of TAK-981 monotherapy in immunocompetent and immunodeficient mouse models using a mouse-derived CMT167 cell line was significant but modest. Since MAPK inhibition of the KRAS downstream pathway is crucial in KRAS-mutant cancer, we expected that co-inhibition of SUMOylation and MEK might be a good option. Surprisingly, combination treatment with TAK-981 and trametinib dramatically induced apoptosis in multiple cell lines and gene-engineered mouse-derived organoids. Moreover, combination therapy resulted in long-term tumor regression in mouse models using cell lines of different tissue types. Finally, we revealed that combination therapy complementally inhibited Rad51 and BRCA1 and accumulated DNA damage. CONCLUSIONS We found that MYC downregulation occurred via SUMOylation inhibition in KRAS-mutant cancer cells. Our findings indicate that dual inhibition of SUMOylation and MEK may be a promising treatment for MYC-expressing KRAS-mutant cancers by enhancing DNA damage accumulation.
Collapse
Affiliation(s)
- Hiroshi Kotani
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan.
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Justin C Boucher
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Tomoyoshi Yamano
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Sakaguchi
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
| | - Shigeki Sato
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
| | - Koji Fukuda
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Akihiro Nishiyama
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
| | - Kaname Yamashita
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
| | - Koushiro Ohtsubo
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
| | - Shinji Takeuchi
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Takumi Nishiuchi
- Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Marco L Davila
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa, 920-0934, Japan.
- Nano Life Science Institute, Kanazawa University, Kanazawa, Japan.
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
20
|
Perrone C, Angioli R, Luvero D, Giannini A, Di Donato V, Cuccu I, Muzii L, Raspagliesi F, Bogani G. Targeting BRAF pathway in low-grade serous ovarian cancer. J Gynecol Oncol 2024; 35:e104. [PMID: 38768941 PMCID: PMC11262891 DOI: 10.3802/jgo.2024.35.e104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/14/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Mutations in genes encoding for proteins along the RAS-RAF-MEK-ERK pathway have been detected in a variety of tumor entities including ovarian carcinomas. In the recent years, several inhibitors of this pathway have been developed, whose antitumor potential is currently being assessed in different clinical trials. Low grade serous ovarian carcinoma, is a rare gynecological tumor which shows favorable overall survival, compared to the general ovarian cancer population, but worrying resistance to conventional chemotherapies. The clinical behavior of low grade serous ovarian carcinoma reflects the different gene profile compared to high-grade serous carcinoma: KRAS/BRAF mutations. BRAF inhibitors as single agents were approved for the treatment of BRAF mutated tumors. Nevertheless, many patients face progressive disease. The understanding of the mechanisms of resistance to BRAF inhibitors therapy and preclinical studies showing that BRAF and mitogen-activated protein kinase kinase (MEK) inhibitors combined therapy delays the onset of resistance compared to BRAF inhibitor single agent, led to the clinical investigation of combined therapy. The aim of this paper is to review the efficacy and safety of the combination of BRAF plus MEK inhibitors on ovarian carcinomas, in particularly focusing on low grade serous ovarian carcinoma.
Collapse
Affiliation(s)
- Chiara Perrone
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberto Angioli
- Department of Gynecology, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Daniela Luvero
- Department of Gynecology, Campus Bio-Medico University Hospital Foundation, Rome, Italy
| | - Andrea Giannini
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Violante Di Donato
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ilaria Cuccu
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Ludovico Muzii
- Department of Gynecological, Obstetrical and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Raspagliesi
- Gynecologic Oncologic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giorgio Bogani
- Gynecologic Oncologic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| |
Collapse
|
21
|
Yoshimura A, Horinaka M, Yaoi T, Ono H, Itoh K, Yamada T, Takayama K, Sakai T. Epithelial-mesenchymal transition status is a remarkable biomarker for the combination treatment with avutometinib and defactinib in KRAS-mutated non-small cell lung cancer. Br J Cancer 2024; 131:361-371. [PMID: 38822146 PMCID: PMC11263561 DOI: 10.1038/s41416-024-02727-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Recent therapeutic strategies for KRAS-mutated cancers that inhibit the MAPK pathway have attracted considerable attention. The RAF/MEK clamp avutometinib (VS-6766/CH5126766/RO5126766/CKI27) is promising for patients with KRAS-mutated cancers. Although avutometinib monotherapy has shown clinical activity in patients with KRAS-mutated cancers, effective combination strategies will be important to develop. METHODS Using a phosphorylation kinase array kit, we explored the feedback mechanism of avutometinib in KRAS-mutated NSCLC cells, and investigated the efficacy of combining avutometinib with inhibitors of the feedback signal using in vitro and in vivo experiments. Moreover, we searched for a biomarker for the efficacy of combination therapy through an in vitro study and analysis using the The Cancer Genome Atlas Programme dataset. RESULTS Focal adhesion kinase (FAK) phosphorylation/activation was increased after avutometinib treatment and synergy between avutometinib and FAK inhibitor, defactinib, was observed in KRAS-mutated NSCLC cells with an epithelial rather than mesenchymal phenotype. Combination therapy with avutometinib and defactinib induced apoptosis with upregulation of Bim in cancer cells with an epithelial phenotype in an in vitro and in vivo study. CONCLUSIONS These results demonstrate that the epithelial-mesenchymal transition status may be a promising biomarker for the efficacy of combination therapy with avutometinib and defactinib in KRAS-mutated NSCLC.
Collapse
Affiliation(s)
- Akihiro Yoshimura
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
- Department of Respiratory Medicine, Japanese Red Cross Kyoto Daini Hospital, 355-5, Haruobi-cho, Kamigyo-ku, Kyoto, 602-8026, Japan
| | - Mano Horinaka
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Takeshi Yaoi
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hisako Ono
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
- Department of Clinical Oncology, Japanese Red Cross Kyoto Daini Hospital, 355-5, Haruobi-cho, Kamigyo-ku, Kyoto, 602-8026, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshiyuki Sakai
- Department of Drug Discovery Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
22
|
Ravix A, Bandiera C, Cardoso E, Lata-Pedreira A, Chtioui H, Decosterd LA, Wagner AD, Schneider MP, Csajka C, Guidi M. Population Pharmacokinetics of Trametinib and Impact of Nonadherence on Drug Exposure in Oncology Patients as Part of the Optimizing Oral Targeted Anticancer Therapies Study. Cancers (Basel) 2024; 16:2193. [PMID: 38927898 PMCID: PMC11201946 DOI: 10.3390/cancers16122193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Trametinib is a targeted therapy used for the treatment of solid tumours, with significant variability reported in real-life studies. This variability increases the risk of suboptimal exposure, which can lead to treatment failure or increased toxicity. Using model-based simulation, this study aims to characterize and investigate the pharmacokinetics and the adequacy of the currently recommended doses of trametinib. Additionally, the simulation of various suboptimal adherence scenarios allowed for an assessment of the impact of patients' drug adherence on the treatment outcome. The population data collected in 33 adult patients, providing 113 plasmatic trametinib concentrations, were best described by a two-compartment model with linear absorption and elimination. The study also identified a significant positive effect of fat-free mass and a negative effect of age on clearance, explaining 66% and 21% of the initial associated variability, respectively. Simulations showed that a maximum dose of 2 mg daily achieved the therapeutic target in 36% of male patients compared to 72% of female patients. A dose of 1.5 mg per day in patients over 65 years of age achieved similar rates, with 44% and 79% for male and female patients, respectively, reaching the therapeutic target. Poor adherence leads to a significant drop in concentrations and a high risk of subtherapeutic drug levels. These results underline the importance of interprofessional collaboration and patient partnership along the patient's journey to address patients' needs regarding trametinib and support medication adherence.
Collapse
Affiliation(s)
- Anne Ravix
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Carole Bandiera
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland (M.P.S.)
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva, Switzerland
- Centre for Primary Care and Public Health (Unisanté), University of Lausanne, 1011 Lausanne, Switzerland
| | - Evelina Cardoso
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland (M.P.S.)
| | - Adrian Lata-Pedreira
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland (M.P.S.)
| | - Haithem Chtioui
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Laurent Arthur Decosterd
- Laboratory of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Anna Dorothea Wagner
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Marie Paule Schneider
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland (M.P.S.)
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva, Switzerland
| | - Chantal Csajka
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland (M.P.S.)
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva, Switzerland
| | - Monia Guidi
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland (M.P.S.)
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
23
|
Shaigany S, Mastacouris N, Tannenbaum R, Strunk A, Luan C, Burshtein J, Burshtein A, Carvajal R, Garg A, Alloo A. Outcome Measurement Instruments Used to Evaluate Dermatologic Adverse Events in Cancer Trials: A Systematic Review. JAMA Dermatol 2024; 160:651-657. [PMID: 38506826 DOI: 10.1001/jamadermatol.2024.0053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Importance Assessment of type, severity, and impact of dermatologic adverse events (DAEs) necessitates well-developed and validated clinician-reported outcome measures (ClinROMs) and patient-reported outcome measures (PROMs) that evaluate concepts specific to mucocutaneous toxic effects and that allow appropriate interpretation and comparison of DAEs across trials. Objective To evaluate heterogeneity and quality of ClinROMs and PROMs used to assess DAEs from systemic cancer therapy. Evidence Review Two systematic reviews were conducted by searching PubMed and Embase databases from inception through March 7, 2023, and April 12, 2023. The first search included randomized clinical trials and observational studies reporting systemic cancer treatment-induced DAEs assessed by a ClinROM or PROM. The second included studies evaluating measurement properties of frequently used ClinROM and PROM instruments. The Consensus-Based Standards for the Selection of Health Measurement Instruments risk of bias tool was used to evaluate methodologic quality of validation assessments. Findings A total of 395 studies were included. The Common Terminology Criteria for Adverse Events (CTCAE) was utilized in 331 studies meeting inclusion criteria (83.8%). At least 1 skin-related PROM was infrequently utilized in systemic chemotherapy clinical trials (79 studies [20.0%]). Most frequently utilized PROMs were the Dermatology Life Quality Index (DLQI; 34 studies [8.6%]) and Skindex-16 (20 studies [5.1%]). Among studies capturing DAEs, 115 (29.1%) reported a nondescript term (ie, rash) as the only DAE. Eight studies described 44 property assessments of the CTCAE, DLQI, and Skindex. There were no studies evaluating content validity, intrarater reliability, or measurement error for the CTCAE, DLQI, or Skindex. There were no studies evaluating structural validity, internal consistency, and responsiveness of DLQI or Skindex. Interrater reliability and responsiveness were each assessed for 1 DAE-related component of the CTCAE. Construct validity for CTCAE, DLQI, and Skindex was evaluated in 29 (65.9%), 3 (6.8%), and 9 (20.5%) assessments, respectively. Conclusions and Relevance In this systematic review, there was a narrow spectrum of ClinROMs and PROMs with limited validity for the measurement of DAEs in the context of systemic chemotherapy interventions in clinical trials. Report of trial DAEs often had low morphologic specificity and meaning. Based on existing gaps in measurement and report of DAEs, a frequent and impactful adverse event to chemotherapy, the framework for evaluating cutaneous toxic effects in oncology trials may need collaborative reevaluation.
Collapse
Affiliation(s)
- Sheila Shaigany
- Northwell Health, New Hyde Park, New York
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
- Northwell Health Cancer Institute, New Hyde Park, New York
| | | | | | | | | | | | | | - Richard Carvajal
- Northwell Health, New Hyde Park, New York
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
- Northwell Health Cancer Institute, New Hyde Park, New York
| | - Amit Garg
- Northwell Health, New Hyde Park, New York
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
- Northwell Health Cancer Institute, New Hyde Park, New York
| | - Allireza Alloo
- Northwell Health, New Hyde Park, New York
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York
- Northwell Health Cancer Institute, New Hyde Park, New York
| |
Collapse
|
24
|
De Carli A, Kapelyukh Y, Kursawe J, Chaplain MAJ, Wolf CR, Hamis S. Simulating BRAFV600E-MEK-ERK signalling dynamics in response to vertical inhibition treatment strategies. NPJ Syst Biol Appl 2024; 10:51. [PMID: 38750040 PMCID: PMC11096323 DOI: 10.1038/s41540-024-00379-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
In vertical inhibition treatment strategies, multiple components of an intracellular pathway are simultaneously inhibited. Vertical inhibition of the BRAFV600E-MEK-ERK signalling pathway is a standard of care for treating BRAFV600E-mutated melanoma where two targeted cancer drugs, a BRAFV600E-inhibitor, and a MEK inhibitor, are administered in combination. Targeted therapies have been linked to early onsets of drug resistance, and thus treatment strategies of higher complexities and lower doses have been proposed as alternatives to current clinical strategies. However, finding optimal complex, low-dose treatment strategies is a challenge, as it is possible to design more treatment strategies than are feasibly testable in experimental settings. To quantitatively address this challenge, we develop a mathematical model of BRAFV600E-MEK-ERK signalling dynamics in response to combinations of the BRAFV600E-inhibitor dabrafenib (DBF), the MEK inhibitor trametinib (TMT), and the ERK-inhibitor SCH772984 (SCH). From a model of the BRAFV600E-MEK-ERK pathway, and a set of molecular-level drug-protein interactions, we extract a system of chemical reactions that is parameterised by in vitro data and converted to a system of ordinary differential equations (ODEs) using the law of mass action. The ODEs are solved numerically to produce simulations of how pathway-component concentrations change over time in response to different treatment strategies, i.e., inhibitor combinations and doses. The model can thus be used to limit the search space for effective treatment strategies that target the BRAFV600E-MEK-ERK pathway and warrant further experimental investigation. The results demonstrate that DBF and DBF-TMT-SCH therapies show marked sensitivity to BRAFV600E concentrations in silico, whilst TMT and SCH monotherapies do not.
Collapse
Affiliation(s)
- Alice De Carli
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK
| | - Yury Kapelyukh
- School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | - Jochen Kursawe
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK
| | - Mark A J Chaplain
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK
| | - C Roland Wolf
- School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | - Sara Hamis
- School of Mathematics and Statistics, University of St Andrews, St Andrews, Scotland, UK.
- Tampere Institute for Advanced Study, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Department of Information Technology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
25
|
Corcoran RB, Do KT, Kim JE, Cleary JM, Parikh AR, Yeku OO, Xiong N, Weekes CD, Veneris J, Ahronian LG, Mauri G, Tian J, Norden BL, Michel AG, Van Seventer EE, Siravegna G, Camphausen K, Chi G, Fetter IJ, Brugge JS, Chen H, Takebe N, Penson RT, Juric D, Flaherty KT, Sullivan RJ, Clark JW, Heist RS, Matulonis UA, Liu JF, Shapiro GI. Phase I/II Study of Combined BCL-xL and MEK Inhibition with Navitoclax and Trametinib in KRAS or NRAS Mutant Advanced Solid Tumors. Clin Cancer Res 2024; 30:1739-1749. [PMID: 38456660 PMCID: PMC11061595 DOI: 10.1158/1078-0432.ccr-23-3135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/11/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE MEK inhibitors (MEKi) lack monotherapy efficacy in most RAS-mutant cancers. BCL-xL is an anti-apoptotic protein identified by a synthetic lethal shRNA screen as a key suppressor of apoptotic response to MEKi. PATIENTS AND METHODS We conducted a dose escalation study (NCT02079740) of the BCL-xL inhibitor navitoclax and MEKi trametinib in patients with RAS-mutant tumors with expansion cohorts for: pancreatic, gynecologic (GYN), non-small cell lung cancer (NSCLC), and other cancers harboring KRAS/NRAS mutations. Paired pretreatment and day 15 tumor biopsies and serial cell-free (cf)DNA were analyzed. RESULTS A total of 91 patients initiated treatment, with 38 in dose escalation. Fifty-eight percent had ≥3 prior therapies. A total of 15 patients (17%) had colorectal cancer, 19 (11%) pancreatic, 15 (17%) NSCLC, and 32 (35%) GYN cancers. The recommended phase II dose (RP2D) was established as trametinib 2 mg daily days 1 to 14 and navitoclax 250 mg daily days 1 to 28 of each cycle. Most common adverse events included diarrhea, thrombocytopenia, increased AST/ALT, and acneiform rash. At RP2D, 8 of 49 (16%) evaluable patients achieved partial response (PR). Disease-specific differences in efficacy were noted. In patients with GYN at the RP2D, 7 of 21 (33%) achieved a PR and median duration of response 8.2 months. No PRs occurred in patients with colorectal cancer, NSCLC, or pancreatic cancer. MAPK pathway inhibition was observed in on-treatment tumor biopsies. Reductions in KRAS/NRAS mutation levels in cfDNA correlated with clinical benefit. CONCLUSIONS Navitoclax in combination with trametinib was tolerable. Durable clinical responses were observed in patients with RAS-mutant GYN cancers, warranting further evaluation in this population.
Collapse
Affiliation(s)
- Ryan B. Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Khanh T. Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jeong E. Kim
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Aparna R. Parikh
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Oladapo O. Yeku
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Niya Xiong
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Colin D. Weekes
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Veneris
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Leanne G. Ahronian
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gianluca Mauri
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, and Department of Hematology Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Jun Tian
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Bryanna L. Norden
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alexa G. Michel
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Emily E. Van Seventer
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Giulia Siravegna
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kyle Camphausen
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gary Chi
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Isobel J. Fetter
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Joan S. Brugge
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Helen Chen
- National Institute of Health, National Cancer Institute, Division of Cancer Treatment and Diagnosis, Cancer Therapy Evaluation Program, Bethesda, Maryland
| | - Naoko Takebe
- National Institute of Health, National Cancer Institute, Division of Cancer Treatment and Diagnosis, Cancer Therapy Evaluation Program, Bethesda, Maryland
| | - Richard T. Penson
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Dejan Juric
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Keith T. Flaherty
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ryan J. Sullivan
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey W. Clark
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Rebecca S. Heist
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ursula A. Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joyce F. Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
26
|
Dreikhausen L, Klupsch A, Wiest I, Xiao Q, Schulte N, Betge J, Boch T, Brochhausen C, Gaiser T, Hofheinz RD, Ebert M, Zhan T. Clinical impact of panel gene sequencing on therapy of advanced cancers of the digestive system: a retrospective, single center study. BMC Cancer 2024; 24:526. [PMID: 38664720 PMCID: PMC11046933 DOI: 10.1186/s12885-024-12261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Panel gene sequencing is an established diagnostic tool for precision oncology of solid tumors, but its utility for the treatment of cancers of the digestive system in clinical routine is less well documented. METHODS We retrospectively identified patients with advanced or metastatic gastrointestinal, pancreaticobiliary or hepatic cancers who received panel gene sequencing at a tertiary university hospital from 2015 to 2022. For these cases, we determined the spectrum of genetic alterations, clinicopathological parameters and treatment courses. Assessment of actionability of genetic alterations was based on the OncoKB database, cancer-specific ESMO treatment guidelines, and recommendations of the local molecular tumor board. RESULTS In total, 155 patients received panel gene sequencing using either the Oncomine Focus (62 cases), Comprehensive (91 cases) or Childhood Cancer Research Assay (2 cases). The mean age of patients was 61 years (range 24-90) and 37% were female. Most patients suffered from either colorectal cancer (53%) or cholangiocellular carcinoma (19%). 327 genetic alterations were discovered in 123 tumor samples, with an average number of 2.1 alterations per tumor. The most frequently altered genes were TP53, KRAS and PIK3CA. Actionable gene alterations were detected in 13.5-56.8% of tumors, according to ESMO guidelines or the OncoKB database, respectively. Thirteen patients were treated with targeted therapies based on identified molecular alterations, with a median progression-free survival of 8.8 months. CONCLUSIONS Actionable genetic alterations are frequently detected by panel gene sequencing in patients with advanced cancers of the digestive tract, providing clinical benefit in selected cases. However, for the majority of identified actionable alterations, sufficient clinical evidence for targeted treatments is still lacking.
Collapse
Affiliation(s)
- Lena Dreikhausen
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna Klupsch
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Isabella Wiest
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Qiyun Xiao
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nadine Schulte
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Betge
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center, Heidelberg, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany
| | - Tobias Boch
- Department of Medicine III, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Timo Gaiser
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ralf-Dieter Hofheinz
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Medicine III, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Medical Faculty Mannheim, DKFZ-Hector Cancer Institute, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
27
|
Kim J, Lee TS, Lee MH, Cho IR, Ryu JK, Kim YT, Lee SH, Paik WH. Pancreatic Cancer Treatment Targeting the HGF/c-MET Pathway: The MEK Inhibitor Trametinib. Cancers (Basel) 2024; 16:1056. [PMID: 38473413 DOI: 10.3390/cancers16051056] [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: 01/31/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Pancreatic cancer is characterized by fibrosis/desmoplasia in the tumor microenvironment, which is primarily mediated by pancreatic stellate cells and cancer-associated fibroblasts. HGF/c-MET signaling, which is instrumental in embryonic development and wound healing, is also implicated for its mitogenic and motogenic properties. In pancreatic cancer, this pathway, along with its downstream signaling pathways, is associated with disease progression, prognosis, metastasis, chemoresistance, and other tumor-related factors. Other features of the microenvironment in pancreatic cancer with the HGF/c-MET pathway include hypoxia, angiogenesis, metastasis, and the urokinase plasminogen activator positive feed-forward loop. All these attributes critically influence the initiation, progression, and metastasis of pancreatic cancer. Therefore, targeting the HGF/c-MET signaling pathway appears promising for the development of innovative drugs for pancreatic cancer treatment. One of the primary downstream effects of c-MET activation is the MAPK/ERK (Ras, Ras/Raf/MEK/ERK) signaling cascade, and MEK (Mitogen-activated protein kinase kinase) inhibitors have demonstrated therapeutic value in RAS-mutant melanoma and lung cancer. Trametinib is a selective MEK1 and MEK2 inhibitor, and it has evolved as a pivotal therapeutic agent targeting the MAPK/ERK pathway in various malignancies, including BRAF-mutated melanoma, non-small cell lung cancer and thyroid cancer. The drug's effectiveness increases when combined with agents like BRAF inhibitors. However, resistance remains a challenge, necessitating ongoing research to counteract the resistance mechanisms. This review offers an in-depth exploration of the HGF/c-MET signaling pathway, trametinib's mechanism, clinical applications, combination strategies, and future directions in the context of pancreatic cancer.
Collapse
Affiliation(s)
- Junyeol Kim
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Tae Seung Lee
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Myeong Hwan Lee
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - In Rae Cho
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Ji Kon Ryu
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yong-Tae Kim
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Woo Hyun Paik
- Department of Internal Medicine, Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
28
|
Hanrahan AJ, Chen Z, Rosen N, Solit DB. BRAF - a tumour-agnostic drug target with lineage-specific dependencies. Nat Rev Clin Oncol 2024; 21:224-247. [PMID: 38278874 PMCID: PMC11857949 DOI: 10.1038/s41571-023-00852-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/28/2024]
Abstract
In June 2022, the FDA granted Accelerated Approval to the BRAF inhibitor dabrafenib in combination with the MEK inhibitor trametinib for the treatment of adult and paediatric patients (≥6 years of age) with unresectable or metastatic BRAFV600E-mutant solid tumours, except for BRAFV600E-mutant colorectal cancers. The histology-agnostic approval of dabrafenib plus trametinib marks the culmination of two decades of research into the landscape of BRAF mutations in human cancers, the biochemical mechanisms underlying BRAF-mediated tumorigenesis, and the clinical development of selective RAF and MEK inhibitors. Although the majority of patients with BRAFV600E-mutant tumours derive clinical benefit from BRAF inhibitor-based combinations, resistance to treatment develops in most. In this Review, we describe the biochemical basis for oncogenic BRAF-induced activation of MAPK signalling and pan-cancer and lineage-specific mechanisms of intrinsic, adaptive and acquired resistance to BRAF inhibitors. We also discuss novel RAF inhibitors and drug combinations designed to delay the emergence of treatment resistance and/or expand the population of patients with BRAF-mutant cancers who benefit from molecularly targeted therapies.
Collapse
Affiliation(s)
- Aphrothiti J Hanrahan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ziyu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
29
|
Cong B, Thakur T, Uribe AH, Stamou E, Gopinath S, Maddocks O, Cagan R. Colon Cancer Cells Evade Drug Action by Enhancing Drug Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572817. [PMID: 38187524 PMCID: PMC10769412 DOI: 10.1101/2023.12.21.572817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Colorectal cancer (CRC) is the second most deadly cancer worldwide. One key reason is the failure of therapies that target RAS proteins, which represent approximately 40% of CRC cases. Despite the recent discovery of multiple alternative signalling pathways that contribute to resistance, durable therapies remain an unmet need. Here, we use liquid chromatography/mass spectrometry (LC/MS) analyses on Drosophila CRC tumour models to identify multiple metabolites in the glucuronidation pathway-a toxin clearance pathway-as upregulated in trametinib-resistant RAS/APC/P53 ("RAP") tumours compared to trametinib-sensitive RASG12V tumours. Elevating glucuronidation was sufficient to direct trametinib resistance in RASG12V animals while, conversely, inhibiting different steps along the glucuronidation pathway strongly reversed RAP resistance to trametinib. For example, blocking an initial HDAC1-mediated deacetylation step with the FDA-approved drug vorinostat strongly suppressed trametinib resistance in Drosophila RAP tumours. We provide functional evidence that pairing oncogenic RAS with hyperactive WNT activity strongly elevates PI3K/AKT/GLUT signalling, which in turn directs elevated glucose and subsequent glucuronidation. Finally, we show that this mechanism of trametinib resistance is conserved in an KRAS/APC/TP53 mouse CRC tumour organoid model. Our observations demonstrate a key mechanism by which oncogenic RAS/WNT activity promotes increased drug clearance in CRC. The majority of targeted therapies are glucuronidated, and our results provide a specific path towards abrogating this resistance in clinical trials.
Collapse
Affiliation(s)
- Bojie Cong
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Teena Thakur
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, Scotland G61 1BD UK
| | - Alejandro Huerta Uribe
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, Scotland G61 1BD UK
| | - Evangelia Stamou
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Sindhura Gopinath
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 25-82 Annenberg Building; Box 1020, One Gustave L. Levy Place, New York, NY 10029
| | - Oliver Maddocks
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Ross Cagan
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| |
Collapse
|
30
|
Cong B, Stamou E, Pennel K, Mckenzie M, Matly A, Gopinath S, Edwards J, Cagan R. WNT Signalling Promotes NF-κB Activation and Drug Resistance in KRAS-Mutant Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572810. [PMID: 38187607 PMCID: PMC10769410 DOI: 10.1101/2023.12.21.572810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Approximately 40% of colorectal cancer (CRC) cases are characterized by KRAS mutations, rendering them insensitive to most CRC therapies. While the reasons for this resistance remain incompletely understood, one key aspect is genetic complexity: in CRC, oncogenic KRAS is most commonly paired with mutations that alter WNT and P53 activities ("RAP"). Here, we demonstrate that elevated WNT activity upregulates canonical (NF-κB) signalling in both Drosophila and human RAS mutant tumours. This upregulation required Toll-1 and Toll-9 and resulted in reduced efficacy of RAS pathway targeted drugs such as the MEK inhibitor trametinib. Inhibiting WNT activity pharmacologically significantly suppressed trametinib resistance in RAP tumours and more genetically complex RAP-containing 'patient avatar' models. WNT/MEK drug inhibitor combinations were further improved by targeting brm, shg, ago, rhoGAPp190 and upf1, highlighting these genes as candidate biomarkers for patients sensitive to this duel approach. These findings shed light on how genetic complexity impacts drug resistance and proposes a therapeutic strategy to reverse this resistance.
Collapse
Affiliation(s)
- Bojie Cong
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Evangelia Stamou
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Kathryn Pennel
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Molly Mckenzie
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Amna Matly
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Sindhura Gopinath
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 25-82 Annenberg Building; Box 1020, One Gustave L. Levy Place, New York, NY 10029
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| | - Ross Cagan
- School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre; Garscube Estate, Switchback Road, Bearsden; Glasgow, Scotland G61 1QH UK
| |
Collapse
|
31
|
Hsu WH, LaBella KA, Lin Y, Xu P, Lee R, Hsieh CE, Yang L, Zhou A, Blecher JM, Wu CJ, Lin K, Shang X, Jiang S, Spring DJ, Xia Y, Chen P, Shen JP, Kopetz S, DePinho RA. Oncogenic KRAS Drives Lipofibrogenesis to Promote Angiogenesis and Colon Cancer Progression. Cancer Discov 2023; 13:2652-2673. [PMID: 37768068 PMCID: PMC10807546 DOI: 10.1158/2159-8290.cd-22-1467] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 08/01/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023]
Abstract
Oncogenic KRAS (KRAS*) contributes to many cancer hallmarks. In colorectal cancer, KRAS* suppresses antitumor immunity to promote tumor invasion and metastasis. Here, we uncovered that KRAS* transforms the phenotype of carcinoma-associated fibroblasts (CAF) into lipid-laden CAFs, promoting angiogenesis and tumor progression. Mechanistically, KRAS* activates the transcription factor CP2 (TFCP2) that upregulates the expression of the proadipogenic factors BMP4 and WNT5B, triggering the transformation of CAFs into lipid-rich CAFs. These lipid-rich CAFs, in turn, produce VEGFA to spur angiogenesis. In KRAS*-driven colorectal cancer mouse models, genetic or pharmacologic neutralization of TFCP2 reduced lipid-rich CAFs, lessened tumor angiogenesis, and improved overall survival. Correspondingly, in human colorectal cancer, lipid-rich CAF and TFCP2 signatures correlate with worse prognosis. This work unveils a new role for KRAS* in transforming CAFs, driving tumor angiogenesis and disease progression, providing an actionable therapeutic intervention for KRAS*-driven colorectal cancer. SIGNIFICANCE This study identified a molecular mechanism contributing to KRAS*-driven colorectal cancer progression via fibroblast transformation in the tumor microenvironment to produce VEGFA driving tumor angiogenesis. In preclinical models, targeting the KRAS*-TFCP2-VEGFA axis impaired tumor progression, revealing a potential novel therapeutic option for patients with KRAS*-driven colorectal cancer. This article is featured in Selected Articles from This Issue, p. 2489.
Collapse
Affiliation(s)
- Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyle A. LaBella
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yiyun Lin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ping Xu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rumi Lee
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cheng-En Hsieh
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lei Yang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ashley Zhou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan M. Blecher
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chang-Jiun Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kangyu Lin
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Jiang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Denise J. Spring
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Xia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peiwen Chen
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A. DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
32
|
Van Cutsem E, Yaeger R, Delord JP, Tabernero J, Siu LL, Ducreux M, Siena S, Elez E, Kasper S, Zander T, Steeghs N, Murphy D, Edwards M, Wainberg ZA. Phase Ib/II Study of the Efficacy and Safety of Binimetinib (MEK162) Plus Panitumumab for Mutant or Wild-Type RAS Metastatic Colorectal Cancer. Oncologist 2023; 28:e1209-e1218. [PMID: 37597246 PMCID: PMC10712701 DOI: 10.1093/oncolo/oyad210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/08/2023] [Indexed: 08/21/2023] Open
Abstract
INTRODUCTION Activating RAS gene mutations occur in approximately 55% of patients with metastatic colorectal cancer (mCRC) and are associated with poorer clinical outcomes due to epidermal growth factor receptor (EGFR) blockade resistance. Combined EGFR and mitogen-activated protein kinase (MEK) inhibition may extend response to EGFR inhibition and overcome acquired resistance. This phase Ib/II dose escalation trial evaluated the safety and activity of dual inhibition with binimetinib (MEK1/2 inhibitor) and panitumumab (EGFR inhibitor [EGFRi]) in patients with RAS mutant or BRAF wild type (WT)/RAS WT mCRC. METHODS Phase Ib dose escalation started with binimetinib 45 mg twice daily plus panitumumab 6 mg/kg administered every 2 weeks. In the phase II study, patients with measurable mCRC were enrolled into 4 groups based on previous anti-EGFR monoclonal antibody therapy and RAS mutational status. RESULTS No patients in the phase Ib portion (n = 10) had a response; 70% of patients had stable disease. In the phase II portion (n = 43), overall response rate (ORR, confirmed) was 2.3% with one partial response in the RAS WT group, DCR was 30.2%, and median progression-free survival was 1.8 months (95%CI, 1.6-3.3). All patients experienced ≥1 adverse event, with the most common being diarrhea (71.7%), vomiting (52.8%), nausea (50.9%), fatigue (49.1%), dermatitis acneiform (43.4%), and rash (41.5%). Most patients required treatment interruption or dose reduction due to difficulties tolerating treatment. CONCLUSIONS The combination of binimetinib and panitumumab had substantial toxicity and limited clinical activity for patients with mutant or WT RAS mCRC, independent of EGFRi treatment history (Trial registration: NCT01927341).
Collapse
Affiliation(s)
- Eric Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg, Leuven and KU Leuven, Leuven, Belgium
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jean-Pierre Delord
- Medical Oncology Department, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | - Josep Tabernero
- Medical Oncology Department, Vall d’Hebron Campus Hospital and Institute of Oncology (VHIO), IOB-Quiron, UVic-UICC, Barcelona, Spain
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Oncology, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elena Elez
- Medical Oncology Department, Vall d’Hebron Campus Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Stefan Kasper
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
| | - Thomas Zander
- Department of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne, University of Cologne, Duesseldorf, Germany
- Department of Internal Medicine I, Gastrointestinal Cancer Group Cologne (GCGC), University Clinic Cologne, Cologne, Germany
| | - Neeltje Steeghs
- Department of Medical Oncology and Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Zev A Wainberg
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
33
|
Pita JM, Raspé E, Coulonval K, Decaussin-Petrucci M, Tarabichi M, Dom G, Libert F, Craciun L, Andry G, Wicquart L, Leteurtre E, Trésallet C, Marlow LA, Copland JA, Durante C, Maenhaut C, Cavaco BM, Dumont JE, Costante G, Roger PP. CDK4 phosphorylation status and rational use for combining CDK4/6 and BRAF/MEK inhibition in advanced thyroid carcinomas. Front Endocrinol (Lausanne) 2023; 14:1247542. [PMID: 37964967 PMCID: PMC10641312 DOI: 10.3389/fendo.2023.1247542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Background CDK4/6 inhibitors (CDK4/6i) have been established as standard treatment against advanced Estrogen Receptor-positive breast cancers. These drugs are being tested against several cancers, including in combinations with other therapies. We identified the T172-phosphorylation of CDK4 as the step determining its activity, retinoblastoma protein (RB) inactivation, cell cycle commitment and sensitivity to CDK4/6i. Poorly differentiated (PDTC) and anaplastic (ATC) thyroid carcinomas, the latter considered one of the most lethal human malignancies, represent major clinical challenges. Several molecular evidence suggest that CDK4/6i could be considered for treating these advanced thyroid cancers. Methods We analyzed by two-dimensional gel electrophoresis the CDK4 modification profile and the presence of T172-phosphorylated CDK4 in a collection of 98 fresh-frozen tissues and in 21 cell lines. A sub-cohort of samples was characterized by RNA sequencing and immunohistochemistry. Sensitivity to CDK4/6i (palbociclib and abemaciclib) was assessed by BrdU incorporation/viability assays. Treatment of cell lines with CDK4/6i and combination with BRAF/MEK inhibitors (dabrafenib/trametinib) was comprehensively evaluated by western blot, characterization of immunoprecipitated CDK4 and CDK2 complexes and clonogenic assays. Results CDK4 phosphorylation was detected in all well-differentiated thyroid carcinomas (n=29), 19/20 PDTC, 16/23 ATC and 18/21 thyroid cancer cell lines, including 11 ATC-derived ones. Tumors and cell lines without phosphorylated CDK4 presented very high p16CDKN2A levels, which were associated with proliferative activity. Absence of CDK4 phosphorylation in cell lines was associated with CDK4/6i insensitivity. RB1 defects (the primary cause of intrinsic CDK4/6i resistance) were not found in 5/7 tumors without detectable phosphorylated CDK4. A previously developed 11-gene expression signature identified the likely unresponsive tumors, lacking CDK4 phosphorylation. In cell lines, palbociclib synergized with dabrafenib/trametinib by completely and permanently arresting proliferation. These combinations prevented resistance mechanisms induced by palbociclib, most notably Cyclin E1-CDK2 activation and a paradoxical stabilization of phosphorylated CDK4 complexes. Conclusion Our study supports further clinical evaluation of CDK4/6i and their combination with anti-BRAF/MEK therapies as a novel effective treatment against advanced thyroid tumors. Moreover, the complementary use of our 11 genes predictor with p16/KI67 evaluation could represent a prompt tool for recognizing the intrinsically CDK4/6i insensitive patients, who are potentially better candidates to immediate chemotherapy.
Collapse
Affiliation(s)
- Jaime M. Pita
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Eric Raspé
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Katia Coulonval
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Maxime Tarabichi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Geneviève Dom
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frederick Libert
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
- BRIGHTCore, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ligia Craciun
- Tumor Bank of the Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guy Andry
- Department of Head & Neck and Thoracic Surgery, Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Laurence Wicquart
- Tumorothèque du Groupement de Coopération Sanitaire-Centre Régional de Référence en Cancérologie (C2RC) de Lille, Lille, France
| | - Emmanuelle Leteurtre
- Department of Pathology, Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Inserm, Centre Hospitalo-Universitaire (CHU) Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Christophe Trésallet
- Department of General and Endocrine Surgery - Pitié-Salpêtrière Hospital, Sorbonne University, Assistance Publique des Hôpitaux de Paris, Paris, France
- Department of Digestive, Bariatric and Endocrine Surgery - Avicenne University Hospital, Paris Nord - Sorbonne University, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Laura A. Marlow
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Cosimo Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Carine Maenhaut
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Branca M. Cavaco
- Molecular Endocrinology Group, Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giuseppe Costante
- Departments of Endocrinology and Medical Oncology, Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pierre P. Roger
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
34
|
Tang H, Ge Y, You T, Li X, Wang Y, Cheng Y, Bai C. A real-world analysis of trametinib in combination with hydroxychloroquine or CDK4/6 inhibitor as third- or later-line therapy in metastatic pancreatic adenocarcinoma. BMC Cancer 2023; 23:958. [PMID: 37817078 PMCID: PMC10563303 DOI: 10.1186/s12885-023-11464-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND There are no standard third-line treatment options for metastatic pancreatic ductal adenocarcinoma (mPDAC). Trametinib in combination with hydroxychloroquine (HCQ) or CDK4/6 inhibitors for pancreatic adenocarcinoma showed promising efficacy in preclinical studies. However, the regimens have not been well examined in patients with mPDAC. METHODS Patients with mPDAC who received the combination of trametinib and HCQ or CDK4/6 inhibitors as third- or later-line therapy were reviewed. The efficacy and prognosis were further analyzed. RESULTS A total of 13 mPDAC patients were enrolled, of whom 8 and 5 patients were treated with trametinib plus HCQ or a CDK4/6 inhibitor (palbociclib or abemaciclib), respectively. All enrolled patients had either KRAS G12D or G12V mutations and had received a median of 3 prior lines of therapy (range, 2-6). The median trametinib treatment duration was 1.4 months. Of the 10 patients with measurable disease, only 1 patient achieved stable disease, and the remaining patients had progressive disease. Moreover, in patients treated with trametinib plus HCQ and a CDK4/6 inhibitor, the median progression-free survival was 2.0 and 2.8 months, respectively, and the median overall survival was 4.2 and 4.7 months, respectively. Moreover, 5 (50%) patients experienced grade 3-4 adverse events in 10 patients with available safety data. CONCLUSIONS The combination of trametinib and HCQ or CDK4/6 inhibitors may not be an effective later-line treatment for mPDAC, and the current preliminary findings need to be confirmed by other studies with larger sample sizes.
Collapse
Affiliation(s)
- Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tingting You
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoyuan Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
35
|
Zhang S, Yao HF, Li H, Su T, Jiang SH, Wang H, Zhang ZG, Dong FY, Yang Q, Yang XM. Transglutaminases are oncogenic biomarkers in human cancers and therapeutic targeting of TGM2 blocks chemoresistance and macrophage infiltration in pancreatic cancer. Cell Oncol (Dordr) 2023; 46:1473-1492. [PMID: 37246171 DOI: 10.1007/s13402-023-00824-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/30/2023] Open
Abstract
PURPOSE Transglutaminases (TGs) are multifunctional enzymes exhibiting transglutaminase crosslinking, as well as atypical GTPase/ATPase and kinase activities. Here, we used an integrated comprehensive analysis to assess the genomic, transcriptomic and immunological landscapes of TGs across cancers. METHODS Gene expression and immune cell infiltration patterns across cancers were obtained from The Cancer Genome Atlas (TCGA) database and Gene Set Enrichment Analysis (GSEA) datasets. Western blotting, immunofluorescence staining, enzyme-linked immunosorbent assays, and orthotopic xenograft models were used to validate our database-derived results. RESULTS We found that the overall expression of TGs (designated as the TG score) is significantly upregulated in multiple cancers and related to a worse patient survival. The expression of TG family members can be regulated through multiple mechanisms at the genetic, epigenetic and transcriptional levels. The expression of transcription factors crucial for epithelial-to-mesenchymal transition (EMT) is commonly correlated with the TG score in many cancer types. Importantly, TGM2 expression displays a close connection with chemoresistance to a wide range of chemotherapeutic drugs. We found that TGM2 expression, F13A1 expression and the overall TG score were positively correlated with the infiltration of immune cells in all cancer types tested. Functional and clinical verification revealed that a higher TGM2 expression is linked with a worse patient survival, an increased IC50 value of gemcitabine, and a higher abundance of tumor-infiltrating macrophages in pancreatic cancer. Mechanistically, we found that increased C-C motif chemokine ligand 2 (CCL2) release mediated by TGM2 contributes to macrophage infiltration into the tumor microenvironment. CONCLUSIONS Our results reveal the relevance and molecular networks of TG genes in human cancers and highlight the importance of TGM2 in pancreatic cancer, which may provide promising directions for immunotherapy and for addressing chemoresistance.
Collapse
Affiliation(s)
- Shan Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Hong-Fei Yao
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200217, People's Republic of China
| | - Hui Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Tong Su
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200011, China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Hao Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Pudong District, Shanghai, 200123, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| | - Fang-Yuan Dong
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, People's Republic of China.
| | - Qin Yang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
36
|
Haynes D, Morgan EE, Chu EY. Cutaneous adverse reactions resulting from targeted cancer therapies: histopathologic and clinical findings. Hum Pathol 2023; 140:129-143. [PMID: 37146945 DOI: 10.1016/j.humpath.2023.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Targeted cancer treatments-designed to interfere with specific molecular signals responsible for tumor survival and progression-have shown benefit over conventional chemotherapies but may lead to diverse cutaneous adverse effects. This review highlights clinically significant dermatologic toxicities and their associated histopathologic findings, resulting from various targeted cancer drugs. Case reports and series, clinical trials, reviews, and meta-analyses are included for analysis and summarized herein. Cutaneous side effects resulting from targeted cancer therapies were reported with incidences as high as 90% for certain medications, and reactions are often predictable based on mechanism(s) of action of a given drug. Common and important reaction patterns included: acneiform eruptions, neutrophilic dermatoses, hand-foot skin reaction, secondary cutaneous malignancies, and alopecia. Clinical and histopathologic recognition of these toxicities remains impactful for patient care.
Collapse
Affiliation(s)
- Dylan Haynes
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eric E Morgan
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Emily Y Chu
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Liu Y, Cheng Y, Huang G, Xia X, Wang X, Tian H. Preclinical characterization of tunlametinib, a novel, potent, and selective MEK inhibitor. Front Pharmacol 2023; 14:1271268. [PMID: 37808191 PMCID: PMC10557067 DOI: 10.3389/fphar.2023.1271268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Background: Aberrant activation of RAS-RAF-MEK-ERK signaling pathway has been implicated in more than one-third of all malignancies. MEK inhibitors are promising therapeutic approaches to target this signaling pathway. Though four MEK inhibitors have been approved by FDA, these compounds possess either limited efficacy or unfavorable PK profiles with toxicity issues, hindering their broadly application in clinic. Our efforts were focused on the design and development of a novel MEK inhibitor, which subsequently led to the discovery of tunlametinib. Methods: This study verified the superiority of tunlametinib over the current MEK inhibitors in preclinical studies. The protein kinase selectivity activity of tunlametinib was evaluated against 77 kinases. Anti-proliferation activity was analyzed using the 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) or (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay. ERK and phospho-ERK levels were evaluated by Western blot analysis. Flow cytometry analysis was employed to investigate cell cycle and arrest. Cell-derived xenograft (CDX) and Patient-derived xenograft (PDX) models were used to evaluate the tumor growth inhibition. The efficacy of tunlametinib as monotherapy treatment was evaluated in KRAS/BRAF mutant or wild type xenograft model. Furthermore, the combination studies of tunlametinib with BRAF/KRASG12C/SHP2 inhibitors or chemotherapeutic agent were conducted by using the cell proliferation assay in vitro and xenograft models in vivo. Results: In vitro, tunlametinib demonstrated high selectivity with approximately 19-fold greater potency against MEK kinase than MEK162, and nearly 10-100-fold greater potency against RAS/RAF mutant cell lines than AZD6244. In vivo, tunlametinib resulted in dramatic tumor suppression and profound inhibition of ERK phosphorylation in tumor tissue. Mechanistic study revealed that tunlametinib induced cell cycle arrest at G0/G1 phase and apoptosis of cells in a dose-proportional manner. In addition, tunlametinib demonstrated a favorable pharmacokinetic profile with dose-proportionality and good oral bioavailability, with minimal drug exposure accumulation. Furthermore, tunlametinib combined with BRAF/KRASG12C/SHP2 inhibitors or docetaxel showed synergistically enhanced response and marked tumor inhibition. Conclusion: Tunlametinib exhibited a promising approach for treating RAS/RAF mutant cancers alone or as combination therapies, supporting the evaluation in clinical trials. Currently, the first-in-human phase 1 study and pivotal clinical trial of tunlametinib as monotherapy have been completed and pivotal trials as combination therapy are ongoing.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongqi Tian
- Shanghai Kechow Pharma, Inc., Shanghai, China
| |
Collapse
|
38
|
Marín A, Al Mamun A, Patel H, Akamatsu H, Ye D, Sudhan DR, Eli L, Marcelain K, Brown BP, Meiler J, Arteaga CL, Hanker AB. Acquired Secondary HER2 Mutations Enhance HER2/MAPK Signaling and Promote Resistance to HER2 Kinase Inhibition in Breast Cancer. Cancer Res 2023; 83:3145-3158. [PMID: 37404061 PMCID: PMC10530374 DOI: 10.1158/0008-5472.can-22-3617] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/23/2023] [Accepted: 06/30/2023] [Indexed: 07/06/2023]
Abstract
HER2 mutations drive the growth of a subset of breast cancers and are targeted with HER2 tyrosine kinase inhibitors (TKI) such as neratinib. However, acquired resistance is common and limits the durability of clinical responses. Most HER2-mutant breast cancers progressing on neratinib-based therapy acquire secondary mutations in HER2. It is unknown whether these secondary HER2 mutations, other than the HER2T798I gatekeeper mutation, are causal to neratinib resistance. Herein, we show that secondary acquired HER2T862A and HER2L755S mutations promote resistance to HER2 TKIs via enhanced HER2 activation and impaired neratinib binding. While cells expressing each acquired HER2 mutation alone were sensitive to neratinib, expression of acquired double mutations enhanced HER2 signaling and reduced neratinib sensitivity. Computational structural modeling suggested that secondary HER2 mutations stabilize the HER2 active state and reduce neratinib binding affinity. Cells expressing double HER2 mutations exhibited resistance to most HER2 TKIs but retained sensitivity to mobocertinib and poziotinib. Double-mutant cells showed enhanced MEK/ERK signaling, which was blocked by combined inhibition of HER2 and MEK. Together, these findings reveal the driver function of secondary HER2 mutations in resistance to HER2 inhibition and provide a potential treatment strategy to overcome acquired resistance to HER2 TKIs in HER2-mutant breast cancer. SIGNIFICANCE HER2-mutant breast cancers acquire secondary HER2 mutations that drive resistance to HER2 tyrosine kinase inhibitors, which can be overcome by combined inhibition of HER2 and MEK.
Collapse
Affiliation(s)
- Arnaldo Marín
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Doctoral Program in Medical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 838045, Chile
- These authors contributed equally: Arnaldo Marin, Abdullah Al Mamun
| | - Abdullah Al Mamun
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- These authors contributed equally: Arnaldo Marin, Abdullah Al Mamun
| | - Hima Patel
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 838045, Chile
| | - Hiroaki Akamatsu
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Current Address: Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | - Dan Ye
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
| | - Dhivya R. Sudhan
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
| | - Lisa Eli
- Puma Biotechnology, Inc., Los Angeles, CA 90024, USA
| | - Katherine Marcelain
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 838045, Chile
| | - Benjamin P. Brown
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Jens Meiler
- Department of Chemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37240, USA
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, 04103, Germany
| | - Carlos L. Arteaga
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ariella B. Hanker
- UT Southwestern Simmons Comprehensive Cancer Center, Dallas, TX 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
39
|
Yeh C, Park W, Yaeger R. KRAS G12D inhibition in pancreatic cancer: Fas expression facilitates immune clearance. Dev Cell 2023; 58:1515-1516. [PMID: 37699334 DOI: 10.1016/j.devcel.2023.08.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023]
Abstract
In an article in this issue of Developmental Cell and in a second paper in Cancer Cell, Mahadevan et al. demonstrate that KrasG12D suppression remodels the immunosuppressive microenvironment of KrasG12D pancreatic cancers, recruits activated CD8+ cytotoxic T cells, and epigenetically upregulates Fas expression in cancer cells, leading to tumor clearance via Fas/FasL-mediated apoptosis.
Collapse
Affiliation(s)
- Celine Yeh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY 10065, USA
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
40
|
Saltos AN, Creelan BC, Tanvetyanon T, Chiappori AA, Antonia SJ, Shafique MR, Ugrenovic-Petrovic M, Sansil S, Neuger A, Ozakinci H, Boyle TA, Kim J, Haura EB, Gray JE. A phase I/IB trial of binimetinib in combination with erlotinib in NSCLC harboring activating KRAS or EGFR mutations. Lung Cancer 2023; 183:107313. [PMID: 37499521 DOI: 10.1016/j.lungcan.2023.107313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Activating mutations in EGFR or KRAS are highly prevalent in NSCLC, share activation of the MAPK pathway and may be amenable to combination therapy to prevent negative feedback activation. METHODS In this phase 1/1B trial, we tested the combination of binimetinib and erlotinib in patients with advanced NSCLC with at least 1 prior line of treatment (unless with activating EGFR mutation which could be treatment-naïve). A subsequent phase 1B expansion accrued patients with either EGFR- or KRAS-mutation using the recommended phase 2 dose (RP2D) from Phase 1. The primary objective was to evaluate the safety of binimetinib plus erlotinib and establish the RP2D. RESULTS 43 patients enrolled (dose-escalation = 23; expansion = 20). 17 harbored EGFR mutation and 22 had KRAS mutation. The RP2D was erlotinib 100 mg daily and binimetinib 15 mg BID × 5 days/week. Common AEs across all doses included diarrhea (69.8%), rash (44.2%), fatigue (32.6%), and nausea (32.6%), and were primarily grade 1/2. Among KRAS mutant patients, 1 (5%) had confirmed partial response and 8 (36%) achieved stable disease as best overall response. Among EGFR mutant patients, 9 were TKI-naïve with 8 (89%) having partial response, and 8 were TKI-pretreated with no partial responses and 1 (13%) stable disease as best overall response. CONCLUSIONS Binimetinib plus erlotinib demonstrated a manageable safety profile and modest efficacy including one confirmed objective response in a KRAS mutant patient. While clinical utility of this specific combination was limited, these results support development of combinations using novel small molecule inhibitors of RAS, selective EGFR- and other MAPK pathway inhibitors, many of which have improved therapeutic indices. CLINICAL TRIAL REGISTRATION NCT01859026.
Collapse
Affiliation(s)
- Andreas N Saltos
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA.
| | - Ben C Creelan
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Tawee Tanvetyanon
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Alberto A Chiappori
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Scott J Antonia
- Center for Cancer Immunotherapy, Duke Cancer Institute, 20 Duke Medicine Cir., Durham, NC 27710, USA
| | - Michael R Shafique
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | | | - Samer Sansil
- Cancer Pharmacokinetics & Pharmacodynamics Core, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Anthony Neuger
- Cancer Pharmacokinetics & Pharmacodynamics Core, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Hilal Ozakinci
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Theresa A Boyle
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| |
Collapse
|
41
|
Chouari T, La Costa FS, Merali N, Jessel MD, Sivakumar S, Annels N, Frampton AE. Advances in Immunotherapeutics in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:4265. [PMID: 37686543 PMCID: PMC10486452 DOI: 10.3390/cancers15174265] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for up to 95% of all pancreatic cancer cases and is the seventh-leading cause of cancer death. Poor prognosis is a result of late presentation, a lack of screening tests and the fact some patients develop resistance to chemotherapy and radiotherapy. Novel therapies like immunotherapeutics have been of recent interest in pancreatic cancer. However, this field remains in its infancy with much to unravel. Immunotherapy and other targeted therapies have yet to yield significant progress in treating PDAC, primarily due to our limited understanding of the disease immune mechanisms and its intricate interactions with the tumour microenvironment (TME). In this review we provide an overview of current novel immunotherapies which have been studied in the field of pancreatic cancer. We discuss their mechanisms, evidence available in pancreatic cancer as well as the limitations of such therapies. We showcase the potential role of combining novel therapies in PDAC, postulate their potential clinical implications and the hurdles associated with their use in PDAC. Therapies discussed with include programmed death checkpoint inhibitors, Cytotoxic T-lymphocyte-associated protein 4, Chimeric Antigen Receptor-T cell therapy, oncolytic viral therapy and vaccine therapies including KRAS vaccines, Telomerase vaccines, Gastrin Vaccines, Survivin-targeting vaccines, Heat-shock protein (HSP) peptide complex-based vaccines, MUC-1 targeting vaccines, Listeria based vaccines and Dendritic cell-based vaccines.
Collapse
Affiliation(s)
- Tarak Chouari
- Hepato-Pancreato-Biliary Department, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK; (T.C.); (F.S.L.C.); (N.M.)
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7WG, UK; (M.-D.J.); (N.A.)
| | - Francesca Soraya La Costa
- Hepato-Pancreato-Biliary Department, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK; (T.C.); (F.S.L.C.); (N.M.)
| | - Nabeel Merali
- Hepato-Pancreato-Biliary Department, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK; (T.C.); (F.S.L.C.); (N.M.)
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7WG, UK; (M.-D.J.); (N.A.)
- The Minimal Access Therapy Training Unit, University of Surrey, Guildford GU2 7WG, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7WG, UK; (M.-D.J.); (N.A.)
| | - Shivan Sivakumar
- Oncology Department and Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham, Birmingham B15 2TT, UK;
| | - Nicola Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7WG, UK; (M.-D.J.); (N.A.)
| | - Adam E. Frampton
- Hepato-Pancreato-Biliary Department, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK; (T.C.); (F.S.L.C.); (N.M.)
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7WG, UK; (M.-D.J.); (N.A.)
- The Minimal Access Therapy Training Unit, University of Surrey, Guildford GU2 7WG, UK
| |
Collapse
|
42
|
Sekiya S, Fukuda J, Yamamura R, Ooshio T, Satoh Y, Kosuge S, Sato R, Hatanaka KC, Hatanaka Y, Mitsuhashi T, Nakamura T, Matsuno Y, Hirano S, Sonoshita M. Drosophila Screening Identifies Dual Inhibition of MEK and AURKB as an Effective Therapy for Pancreatic Ductal Adenocarcinoma. Cancer Res 2023; 83:2704-2715. [PMID: 37378549 DOI: 10.1158/0008-5472.can-22-3762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023]
Abstract
Significant progress has been made in understanding the pathogenesis of pancreatic ductal adenocarcinoma (PDAC) by generating and using murine models. To accelerate drug discovery by identifying novel therapeutic targets on a systemic level, here we generated a Drosophila model mimicking the genetic signature in PDAC (KRAS, TP53, CDKN2A, and SMAD4 alterations), which is associated with the worst prognosis in patients. The '4-hit' flies displayed epithelial transformation and decreased survival. Comprehensive genetic screening of their entire kinome revealed kinases including MEK and AURKB as therapeutic targets. Consistently, a combination of the MEK inhibitor trametinib and the AURKB inhibitor BI-831266 suppressed the growth of human PDAC xenografts in mice. In patients with PDAC, the activity of AURKB was associated with poor prognosis. This fly-based platform provides an efficient whole-body approach that complements current methods for identifying therapeutic targets in PDAC. SIGNIFICANCE Development of a Drosophila model mimicking genetic alterations in human pancreatic ductal adenocarcinoma provides a tool for genetic screening that identifies MEK and AURKB inhibition as a potential treatment strategy.
Collapse
Affiliation(s)
- Sho Sekiya
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Junki Fukuda
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Takako Ooshio
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yusuke Satoh
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Kosuge
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Reo Sato
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Tomoko Mitsuhashi
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Masahiro Sonoshita
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
| |
Collapse
|
43
|
Aaroe A, Kurzrock R, Goyal G, Goodman AM, Patel H, Ruan G, Ulaner G, Young J, Li Z, Dustin D, Go RS, Diamond EL, Janku F. Successful treatment of non-Langerhans cell histiocytosis with the MEK inhibitor trametinib: a multicenter analysis. Blood Adv 2023; 7:3984-3992. [PMID: 36857436 PMCID: PMC10410131 DOI: 10.1182/bloodadvances.2022009013] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/25/2023] [Accepted: 02/17/2023] [Indexed: 03/03/2023] Open
Abstract
Erdheim-Chester disease (ECD) and Rosai-Dorfman disease (RDD) are rare non-Langerhans cell histiocytoses (non-LCHs), for which therapeutic options are limited. MAPK pathway activation through BRAFV600E mutation or other genomic alterations is a histiocytosis hallmark and correlates with a favorable response to BRAF inhibitors and the MEK inhibitor cobimetinib. However, there has been no systematic evaluation of alternative MEK inhibitors. To assess the efficacy and safety of the MEK inhibitor trametinib, we retrospectively analyzed the outcomes of 26 adult patients (17 with ECD, 5 with ECD/RDD, 3 with RDD, and 1 with ECD/LCH) treated with orally administered trametinib at 4 major US care centers. The most common treatment-related toxicity was rash (27% of patients). In most patients, the disease was effectively managed at low doses (0.5-1.0 mg trametinib daily). The response rate of the 17 evaluable patients was 71% (73% [8/11] without a detectable BRAFV600E achieving response). At a median follow-up of 23 months, treatment effects were durable, with a median time-to-treatment failure of 37 months, whereas the median progression-free and overall survival were not reached (at 3 years, 90.1% of patients were alive). Most patients harbored mutations in BRAF (either classic BRAFV600E or other BRAF alterations) or alterations in other genes involved in the MAPK pathway, eg, MAP2K, NF1, GNAS, or RAS. Most patients required lower than standard doses of trametinib but were responsive to lower doses. Our data suggest that the MEK inhibitor trametinib is an effective treatment for ECD and RDD, including those without the BRAFV600E mutation.
Collapse
Affiliation(s)
- Ashley Aaroe
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, Paris, France
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Gaurav Goyal
- Division of Hematology-Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Aaron M. Goodman
- Division of Hematology and Oncology, University of California San Diego, La Jolla, CA
| | - Harsh Patel
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Gordon Ruan
- Division of Hematology and Department of Radiology, Mayo Clinic, Rochester, MN
| | - Gary Ulaner
- Hoag Family Cancer Institute, Newport Beach, CA
| | - Jason Young
- Division of Hematology and Department of Radiology, Mayo Clinic, Rochester, MN
| | - Ziyi Li
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Derek Dustin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ronald S. Go
- Division of Hematology and Department of Radiology, Mayo Clinic, Rochester, MN
| | - Eli L. Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
44
|
Piha-Paul SA, Tseng C, Tran HT, Gao M, Karp DD, Subbiah V, Tsimberidou AM, Kawedia JD, Fu S, Pant S, Yap TA, Morris VK, Kee BK, Blum Murphy M, Lim J, Meric-Bernstam F. A phase I trial of the pan-ERBB inhibitor neratinib combined with the MEK inhibitor trametinib in patients with advanced cancer with EGFR mutation/amplification, HER2 mutation/amplification, HER3/4 mutation or KRAS mutation. Cancer Chemother Pharmacol 2023; 92:107-118. [PMID: 37314501 PMCID: PMC10326142 DOI: 10.1007/s00280-023-04545-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023]
Abstract
PURPOSE Aberrant alterations of ERBB receptor tyrosine kinases lead to tumorigenesis. Single agent therapy targeting EGFR or HER2 has shown clinical successes, but drug resistance often develops due to aberrant or compensatory mechanisms. Herein, we sought to determine the feasibility and safety of neratinib and trametinib in patients with EGFR mutation/amplification, HER2 mutation/amplification, HER3/4 mutation and KRAS mutation. METHODS Patients with actionable somatic mutations or amplifications in ERBB genes or actionable KRAS mutations were enrolled to receive neratinib and trametinib in this phase I dose escalation trial. The primary endpoint was determination of the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT). Secondary endpoints included pharmacokinetic analysis and preliminary anti-tumor efficacy. RESULTS Twenty patients were enrolled with a median age of 50.5 years and a median of 3 lines of prior therapy. Grade 3 treatment-related toxicities included: diarrhea (25%), vomiting (10%), nausea (5%), fatigue (5%) and malaise (5%). The MTD was dose level (DL) minus 1 (neratinib 160 mg daily with trametinib 1 mg, 5 days on and 2 days off) given 2 DLTs of grade 3 diarrhea in DL1 (neratinib 160 mg daily with trametinib 1 mg daily). The treatment-related toxicities of DL1 included: diarrhea (100%), nausea (55.6%) and rash (55.6%). Pharmacokinetic data showed trametinib clearance was significantly reduced leading to high drug exposures of trametinib. Two patients achieved stable disease (SD) ≥ 4 months. CONCLUSION Neratinib and trametinib combination was toxic and had limited clinical efficacy. This may be due to suboptimal drug dosing given drug-drug interactions. TRIAL REGISTRATION ID NCT03065387.
Collapse
Affiliation(s)
- Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA.
| | - Chieh Tseng
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
| | - Hai T Tran
- Department of Thoracic, Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meng Gao
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
| | - Apostolia Maria Tsimberidou
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
| | - Jitesh D Kawedia
- Pharmacy Pharmacology Research, Division of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
- Department of Thoracic, Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van K Morris
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan K Kee
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariela Blum Murphy
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - JoAnn Lim
- Pharmacy Clinical Programs, Division of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 455, Houston, TX, 77030, USA
- Department of Breast Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
- The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
45
|
Han JW, Chang HS, Yang JY, Choi HS, Park HS, Jun HO, Choi JH, Paik SS, Chung KH, Shin HJ, Nam S, Son JH, Lee SH, Lee EJ, Seo KY, Lyu J, Kim JW, Kim IB, Park TK. Intravitreal Administration of Retinal Organoids-Derived Exosomes Alleviates Photoreceptor Degeneration in Royal College of Surgeons Rats by Targeting the Mitogen-Activated Protein Kinase Pathway. Int J Mol Sci 2023; 24:12068. [PMID: 37569444 PMCID: PMC10419150 DOI: 10.3390/ijms241512068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Increasing evidence suggests that exosomes are involved in retinal cell degeneration, including their insufficient release; hence, they have become important indicators of retinopathies. The exosomal microRNA (miRNA), in particular, play important roles in regulating ocular and retinal cell functions, including photoreceptor maturation, maintenance, and visual function. Here, we generated retinal organoids (ROs) from human induced pluripotent stem cells that differentiated in a conditioned medium for 60 days, after which exosomes were extracted from ROs (Exo-ROs). Subsequently, we intravitreally injected the Exo-RO solution into the eyes of the Royal College of Surgeons (RCS) rats. Intravitreal Exo-RO administration reduced photoreceptor apoptosis, prevented outer nuclear layer thinning, and preserved visual function in RCS rats. RNA sequencing and miRNA profiling showed that exosomal miRNAs are mainly involved in the mitogen-activated protein kinase (MAPK) signaling pathway. In addition, the expression of MAPK-related genes and proteins was significantly decreased in the Exo-RO-treated group. These results suggest that Exo-ROs may be a potentially novel strategy for delaying retinal degeneration by targeting the MAPK signaling pathway.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.W.H.); (H.S.C.); (H.S.P.); (S.H.L.)
| | - Hun Soo Chang
- Department of Microbiolo and BK21 FOUR Project, Soonchunhyang University College of Medicine, Cheonan 31538, Republic of Korea; (H.S.C.); (J.-H.S.)
| | - Jin Young Yang
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.Y.Y.); (H.O.J.); (J.H.C.); (K.H.C.)
| | - Han Sol Choi
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.W.H.); (H.S.C.); (H.S.P.); (S.H.L.)
| | - Hyo Song Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.W.H.); (H.S.C.); (H.S.P.); (S.H.L.)
| | - Hyoung Oh Jun
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.Y.Y.); (H.O.J.); (J.H.C.); (K.H.C.)
| | - Ji Hye Choi
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.Y.Y.); (H.O.J.); (J.H.C.); (K.H.C.)
| | - Sun-Sook Paik
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea; (S.-S.P.); (I.-B.K.)
- Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| | - Kyung Hwun Chung
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.Y.Y.); (H.O.J.); (J.H.C.); (K.H.C.)
| | - Hee Jeong Shin
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea;
| | - Seungyeon Nam
- Department of Neuroscience and Behavior, University of Notre Dame College of Science, Notre Dame, IN 46556, USA;
| | - Ji-Hye Son
- Department of Microbiolo and BK21 FOUR Project, Soonchunhyang University College of Medicine, Cheonan 31538, Republic of Korea; (H.S.C.); (J.-H.S.)
| | - Si Hyung Lee
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.W.H.); (H.S.C.); (H.S.P.); (S.H.L.)
| | - Eun Jung Lee
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; (E.J.L.); (J.W.K.)
| | - Kyoung Yul Seo
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jungmook Lyu
- Department of Medical Science, Konyang University, Daejun 32992, Republic of Korea;
| | - Jin Woo Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; (E.J.L.); (J.W.K.)
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea; (S.-S.P.); (I.-B.K.)
- Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.W.H.); (H.S.C.); (H.S.P.); (S.H.L.)
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea; (J.Y.Y.); (H.O.J.); (J.H.C.); (K.H.C.)
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 31538, Republic of Korea;
- oligoNgene Pharmaceutical Co., Ltd., Bucheon 31538, Republic of Korea
| |
Collapse
|
46
|
Bteich F, Mohammadi M, Li T, Bhat MA, Sofianidi A, Wei N, Kuang C. Targeting KRAS in Colorectal Cancer: A Bench to Bedside Review. Int J Mol Sci 2023; 24:12030. [PMID: 37569406 PMCID: PMC10418782 DOI: 10.3390/ijms241512030] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/21/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with a myriad of alterations at the cellular and molecular levels. Kristen rat sarcoma (KRAS) mutations occur in up to 40% of CRCs and serve as both a prognostic and predictive biomarker. Oncogenic mutations in the KRAS protein affect cellular proliferation and survival, leading to tumorigenesis through RAS/MAPK pathways. Until recently, only indirect targeting of the pathway had been investigated. There are now several KRAS allele-specific inhibitors in late-phase clinical trials, and many newer agents and targeting strategies undergoing preclinical and early-phase clinical testing. The adequate treatment of KRAS-mutated CRC will inevitably involve combination therapies due to the existence of robust adaptive resistance mechanisms in these tumors. In this article, we review the most recent understanding and findings related to targeting KRAS mutations in CRC, mechanisms of resistance to KRAS inhibitors, as well as evolving treatment strategies for KRAS-mutated CRC patients.
Collapse
Affiliation(s)
- Fernand Bteich
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY 10467, USA;
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.); (T.L.); (M.A.B.); (N.W.)
| | - Mahshid Mohammadi
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.); (T.L.); (M.A.B.); (N.W.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Terence Li
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.); (T.L.); (M.A.B.); (N.W.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Muzaffer Ahmed Bhat
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.); (T.L.); (M.A.B.); (N.W.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Amalia Sofianidi
- Oncology Unit, Third Department of Internal Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Ning Wei
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.); (T.L.); (M.A.B.); (N.W.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chaoyuan Kuang
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY 10467, USA;
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (M.M.); (T.L.); (M.A.B.); (N.W.)
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
47
|
Cuevas-Navarro A, Wagner M, Van R, Swain M, Mo S, Columbus J, Allison MR, Cheng A, Messing S, Turbyville TJ, Simanshu DK, Sale MJ, McCormick F, Stephen AG, Castel P. RAS-dependent RAF-MAPK hyperactivation by pathogenic RIT1 is a therapeutic target in Noonan syndrome-associated cardiac hypertrophy. SCIENCE ADVANCES 2023; 9:eadf4766. [PMID: 37450595 PMCID: PMC10348673 DOI: 10.1126/sciadv.adf4766] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
RIT1 is a RAS guanosine triphosphatase (GTPase) that regulates different aspects of signal transduction and is mutated in lung cancer, leukemia, and in the germline of individuals with Noonan syndrome. Pathogenic RIT1 proteins promote mitogen-activated protein kinase (MAPK) hyperactivation; however, this mechanism remains poorly understood. Here, we show that RAF kinases are direct effectors of membrane-bound mutant RIT1 necessary for MAPK activation. We identify critical residues in RIT1 that facilitate interaction with membrane lipids and show that these are necessary for association with RAF kinases and MAPK activation. Although mutant RIT1 binds to RAF kinases directly, it fails to activate MAPK signaling in the absence of classical RAS proteins. Consistent with aberrant RAF/MAPK activation as a driver of disease, we show that pathway inhibition alleviates cardiac hypertrophy in a mouse model of RIT1 mutant Noonan syndrome. These data shed light on the function of pathogenic RIT1 and identify avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Antonio Cuevas-Navarro
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Morgan Wagner
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Richard Van
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Monalisa Swain
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Stephanie Mo
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - John Columbus
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Madeline R. Allison
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alice Cheng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Simon Messing
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Thomas J. Turbyville
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Dhirendra K. Simanshu
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Matthew J. Sale
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Frank McCormick
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andrew G. Stephen
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, MD 21702, USA
| | - Pau Castel
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
48
|
Hu X, Li W, Zeng K, Xu Z, Li C, Kang Z, Li S, Huang X, Han P, Lin H, Hui AM, Tan Y, Diao L, Li B, Wang X, Wu Z, Lin X. Phase 1 dose-escalation study to evaluate the safety, tolerability, pharmacokinetics, and anti-tumor activity of FCN-159 in adults with neurofibromatosis type 1-related unresectable plexiform neurofibromas. BMC Med 2023; 21:230. [PMID: 37400844 DOI: 10.1186/s12916-023-02927-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/07/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Surgery is a common treatment strategy for patients with neurofibromatosis type 1 (NF1)-related plexiform neurofibroma (PN) and has limited efficacy. FCN-159 is a novel anti-tumorigenic drug via selective inhibition of MEK1/2. This study assesses the safety and efficacy of FCN-159 in patients with NF1-related PN. METHODS This is a multicenter, open-label, single-arm, phase I dose-escalation study. Patients with NF1-related PN that was non-resectable or unsuitable for surgery were enrolled; they received FCN-159 monotherapy daily in 28-day cycles. RESULTS Nineteen adults were enrolled in the study, 3 in 4 mg, 4 in 6 mg, 8 in 8 mg, and 4 in 12 mg. Among patients included in dose-limiting toxicity (DLT) analysis, DLTs (grade 3 folliculitis) were reported in 1 of 8 patients (16.7%) receiving 8 mg and 3 of 3 (100%) patients receiving 12 mg. The maximum tolerated dose was determined to be 8 mg. FCN-159-related treatment-emergent adverse events (TEAEs) were observed in 19 patients (100%); most of which were grade 1 or 2. Nine (47.4%) patients reported grade 3 study-drug-related TEAEs across all dose levels, including four experiencing paronychia and five experiencing folliculitis. Of the 16 patients analyzed, all (100%) had reduced tumor size and six (37.5%) achieved partial responses; the largest reduction in tumor size was 84.2%. The pharmacokinetic profile was approximately linear between 4 and 12 mg, and the half-life supported once daily dosing. CONCLUSIONS FCN-159 was well tolerated up to 8 mg daily with manageable adverse events and showed promising anti-tumorigenic activity in patients with NF1-related PN, warranting further investigation in this indication. TRIAL REGISTRATION ClinicalTrials.gov, NCT04954001. Registered 08 July 2021.
Collapse
Affiliation(s)
- Xiaojie Hu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Kang Zeng
- Department of Dermatology, NanFang Hospital Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Changxing Li
- Department of Dermatology, NanFang Hospital Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Zhuang Kang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Shenglan Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Xin Huang
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Pu Han
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Hongmei Lin
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Ai-Min Hui
- Fosun Pharma USA Inc., 91 Hartwell Ave Suite 305, Lexington, MA, 02421, USA
- EnCureGen Pharma, 9 Yayingshi Road, Guangzhou, 510525, China
| | - Yan Tan
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Lei Diao
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Ben Li
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Xingli Wang
- Shanghai Fosun Pharmaceutical Development Co., Ltd, 1289 Yishan Road, Shanghai, 20033, China
| | - Zhuli Wu
- Shanghai Fosun Pharmaceutical Development Co., Ltd, 1289 Yishan Road, Shanghai, 20033, China.
| | - Xiaoxi Lin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
49
|
Cucci MA, Grattarola M, Monge C, Roetto A, Barrera G, Caputo E, Dianzani C, Pizzimenti S. Nrf2 as a Therapeutic Target in the Resistance to Targeted Therapies in Melanoma. Antioxidants (Basel) 2023; 12:1313. [PMID: 37372043 DOI: 10.3390/antiox12061313] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The use of specific inhibitors towards mutant BRAF (BRAFi) and MEK (MEKi) in BRAF-mutated patients has significantly improved progression-free and overall survival of metastatic melanoma patients. Nevertheless, half of the patients still develop resistance within the first year of therapy. Therefore, understanding the mechanisms of BRAFi/MEKi-acquired resistance has become a priority for researchers. Among others, oxidative stress-related mechanisms have emerged as a major force. The aim of this study was to evaluate the contribution of Nrf2, the master regulator of the cytoprotective and antioxidant response, in the BRAFi/MEKi acquired resistance of melanoma. Moreover, we investigated the mechanisms of its activity regulation and the possible cooperation with the oncogene YAP, which is also involved in chemoresistance. Taking advantage of established in vitro melanoma models resistant to BRAFi, MEKi, or dual resistance to BRAFi/MEKi, we demonstrated that Nrf2 was upregulated in melanoma cells resistant to targeted therapy at the post-translational level and that the deubiquitinase DUB3 participated in the control of the Nrf2 protein stability. Furthermore, we found that Nrf2 controlled the expression of YAP. Importantly, the inhibition of Nrf2, directly or through inhibition of DUB3, reverted the resistance to targeted therapies.
Collapse
Affiliation(s)
- Marie Angèle Cucci
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Margherita Grattarola
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Chiara Monge
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Sciences-San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Emilia Caputo
- Institute of Genetics and Biophysics-IGB-CNR, "A. Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| |
Collapse
|
50
|
Piringer G, Decker J, Trommet V, Kühr T, Heibl S, Dörfler K, Thaler J. Ongoing complete response after treatment cessation with dabrafenib, trametinib, and cetuximab as third-line treatment in a patient with advanced BRAF V600E mutated, microsatellite-stable colon cancer: A case report and literature review. Front Oncol 2023; 13:1166545. [PMID: 37213293 PMCID: PMC10196488 DOI: 10.3389/fonc.2023.1166545] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/28/2023] [Indexed: 05/23/2023] Open
Abstract
Metastatic BRAFV600E mutated colorectal cancer is associated with poor overall survival and modest effectiveness to standard therapies. Furthermore, survival is influenced by the microsatellite status. Patients with microsatellite-stable and BRAFV600E mutated colorectal cancer have the worst prognosis under the wide range of genetic subgroups in colorectal cancer. Herein, we present a patient case of an impressive therapeutic efficacy of dabrafenib, trametinib, and cetuximab as later-line therapy in a 52-year-old woman with advanced BRAFV600E mutated, microsatellite-stable colon cancer. This patient achieved a complete response after 1 year of triple therapy. Due to skin toxicity grade 3 and recurrent urinary tract infections due to mucosal toxicity, a therapy de-escalation to dabrafenib and trametinib was performed, and the double therapy was administered for further 41 months with ongoing complete response. For 1 year, the patient was off therapy and is still in complete remission.
Collapse
Affiliation(s)
- Gudrun Piringer
- Department of Internal Medicine IV, Wels-Grieskirchen Medical Hospital, Wels, Austria
- Department of Hematology and Oncology, Kepler University Hospital, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Jörn Decker
- Department of Internal Medicine, Klinikum Rohrbach, Rohrbach, Austria
| | - Vera Trommet
- Department of Internal Medicine IV, Wels-Grieskirchen Medical Hospital, Wels, Austria
| | - Thomas Kühr
- Department of Internal Medicine IV, Wels-Grieskirchen Medical Hospital, Wels, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Sonja Heibl
- Department of Internal Medicine IV, Wels-Grieskirchen Medical Hospital, Wels, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Konrad Dörfler
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Josef Thaler
- Department of Internal Medicine IV, Wels-Grieskirchen Medical Hospital, Wels, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|