51
|
Pulumati A, Pulumati A, Dwarakanath BS, Verma A, Papineni RVL. Technological advancements in cancer diagnostics: Improvements and limitations. Cancer Rep (Hoboken) 2023; 6:e1764. [PMID: 36607830 PMCID: PMC9940009 DOI: 10.1002/cnr2.1764] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/20/2022] [Accepted: 11/27/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Cancer is characterized by the rampant proliferation, growth, and infiltration of malignantly transformed cancer cells past their normal boundaries into adjacent tissues. It is the leading cause of death worldwide, responsible for approximately 19.3 million new diagnoses and 10 million deaths globally in 2020. In the United States alone, the estimated number of new diagnoses and deaths is 1.9 million and 609 360, respectively. Implementation of currently existing cancer diagnostic techniques such as positron emission tomography (PET), X-ray computed tomography (CT), and magnetic resonance spectroscopy (MRS), and molecular diagnostic techniques, have enabled early detection rates and are instrumental not only for the therapeutic management of cancer patients, but also for early detection of the cancer itself. The effectiveness of these cancer screening programs are heavily dependent on the rate of accurate precursor lesion identification; an increased rate of identification allows for earlier onset treatment, thus decreasing the incidence of invasive cancer in the long-term, and improving the overall prognosis. Although these diagnostic techniques are advantageous due to lack of invasiveness and easier accessibility within the clinical setting, several limitations such as optimal target definition, high signal to background ratio and associated artifacts hinder the accurate diagnosis of specific types of deep-seated tumors, besides associated high cost. In this review we discuss various imaging, molecular, and low-cost diagnostic tools and related technological advancements, to provide a better understanding of cancer diagnostics, unraveling new opportunities for effective management of cancer, particularly in low- and middle-income countries (LMICs). RECENT FINDINGS Herein we discuss various technological advancements that are being utilized to construct an assortment of new diagnostic techniques that incorporate hardware, image reconstruction software, imaging devices, biomarkers, and even artificial intelligence algorithms, thereby providing a reliable diagnosis and analysis of the tumor. Also, we provide a brief account of alternative low cost-effective cancer therapy devices (CryoPop®, LumaGEM®, MarginProbe®) and picture archiving and communication systems (PACS), emphasizing the need for multi-disciplinary collaboration among radiologists, pathologists, and other involved specialties for improving cancer diagnostics. CONCLUSION Revolutionary technological advancements in cancer imaging and molecular biology techniques are indispensable for the accurate diagnosis and prognosis of cancer.
Collapse
Affiliation(s)
- Akhil Pulumati
- University of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Anika Pulumati
- University of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Bilikere S. Dwarakanath
- Central Research FacilitySri Ramachandra Institute of Higher Education and Research PorurChennaiIndia
- Department of BiotechnologyIndian Academy Degree CollegeBangaloreIndia
| | | | - Rao V. L. Papineni
- PACT & Health LLCBranfordConnecticutUSA
- Department of SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| |
Collapse
|
52
|
Zhou J, Cipriani A, Liu Y, Fang G, Li Q, Cao Y. Mapping lesion-specific response and progression dynamics and inter-organ variability in metastatic colorectal cancer. Nat Commun 2023; 14:417. [PMID: 36697416 PMCID: PMC9876906 DOI: 10.1038/s41467-023-36121-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Achieving systemic tumor control across metastases is vital for long-term patient survival but remains intractable in many patients. High lesion-level response heterogeneity persists, conferring many dissociated responses across metastatic lesions. Most studies of metastatic disease focus on tumor molecular and cellular features, which are crucial to elucidating the mechanisms underlying lesion-level variability. However, our understanding of lesion-specific heterogeneity on the macroscopic level, such as lesion dynamics in growth, response, and progression during treatment, remains rudimentary. This study investigates lesion-specific response heterogeneity through analyzing 116,542 observations of 40,612 lesions in 4,308 metastatic colorectal cancer (mCRC) patients. Despite significant differences in their response and progression dynamics, metastatic lesions converge on four phenotypes that vary with anatomical site. Importantly, we find that organ-level progression sequence is closely associated with patient long-term survival, and that patients with the first lesion progression in the liver often have worse survival. In conclusion, our study provides insights into lesion-specific response and progression heterogeneity in mCRC and creates impetus for metastasis-specific therapeutics.
Collapse
Affiliation(s)
- Jiawei Zhou
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Amber Cipriani
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Health Medical Center, Department of Pharmacy, Chapel Hill, NC, 27514, USA
| | - Yutong Liu
- School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Gang Fang
- Division of Pharmaceutical Outcomes and Policy, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Quefeng Li
- School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
53
|
Jia LL, Zhao JX, Zhao LP, Tian JH, Huang G. Current status and quality of radiomic studies for predicting KRAS mutations in colorectal cancer patients: A systematic review and meta‑analysis. Eur J Radiol 2023; 158:110640. [PMID: 36525703 DOI: 10.1016/j.ejrad.2022.110640] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/13/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE The purpose of this study was to evaluate the methodological quality of radiomics-based studies for noninvasive, preoperative prediction of Kirsten rat sarcoma (KRAS) mutations in patients with colorectal cancer; furthermore, we systematically evaluate the diagnostic accuracy of predicting models. METHODS We systematically searched PubMed, Embase, Cochrane Library and Web of Science databases up to 20 April 2022 for eligible studies. The methodological quality of included studies was assessed using the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2) and Radiomics Quality Score (RQS) tools. A meta-analysis of studies on the prediction of KRAS status in colorectal cancer patients was performed. RESULT Twenty-nine studies were identified in the systematic review, including three studies on the prediction of KRAS status in colorectal cancer liver metastases. All studies had an average RQS score of 9.55 (26.5% of the total score), ranging from 3 to 17. Most studies demonstrated a low or unclear risk of bias in the domains of QUADAS-2. Nineteen studies were included in the meta-analysis, mostly imaged with magnetic resonance imaging (MRI), followed by computed tomography (CT), positron emission tomography-CT (PET/CT). With pooled sensitivity, specificity and area under the curve (AUC) of the training cohorts were 0.80(95% confidence interval(CI), 0.75-0.84), 0.80(95% CI, 0.74-0.85) and 0.87(95% CI, 0.84-0.90),respectively. The pooled sensitivity, specificity, and AUC for the validation cohorts (13 studies) were 0.78(95% CI, 0.71-0.84), 0.84(95% CI, 0.74-0.90), and 0.86(95% CI, 0.83-0.89), respectively. CONCLUSION Radiomics is a potential noninvasive technology that has a moderate preoperative diagnosis and prediction effect on KRAS mutations. However, it has not been implemented as a clinical decision-making tool. Future researchers should pay more attention to the methodological quality of the study and further externally validate the model using multicenter datasets.
Collapse
Affiliation(s)
- Lu-Lu Jia
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou 73000, China.
| | - Jian-Xin Zhao
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou 73000, China.
| | - Lian-Ping Zhao
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China.
| | - Jin-Hui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Gang Huang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou 730000, China.
| |
Collapse
|
54
|
Telekes A, Horváth A. The Role of Cell-Free DNA in Cancer Treatment Decision Making. Cancers (Basel) 2022; 14:6115. [PMID: 36551600 PMCID: PMC9776613 DOI: 10.3390/cancers14246115] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The aim of this review is to evaluate the present status of the use of cell-free DNA and its fraction of circulating tumor DNA (ctDNA) because this year July 2022, an ESMO guideline was published regarding the application of ctDNA in patient care. This review is for clinical oncologists to explain the concept, the terms used, the pros and cons of ctDNA; thus, the technical aspects of the different platforms are not reviewed in detail, but we try to help in navigating the current knowledge in liquid biopsy. Since the validated and adequately sensitive ctDNA assays have utility in identifying actionable mutations to direct targeted therapy, ctDNA may be used for this soon in routine clinical practice and in other different areas as well. The cfDNA fragments can be obtained by liquid biopsy and can be used for diagnosis, prognosis, and selecting among treatment options in cancer patients. A great proportion of cfDNA comes from normal cells of the body or from food uptake. Only a small part (<1%) of it is related to tumors, originating from primary tumors, metastatic sites, or circulating tumor cells (CTCs). Soon the data obtained from ctDNA may routinely be used for finding minimal residual disease, detecting relapse, and determining the sites of metastases. It might also be used for deciding appropriate therapy, and/or emerging resistance to the therapy and the data analysis of ctDNA may be combined with imaging or other markers. However, to achieve this goal, further clinical validations are inevitable. As a result, clinicians should be aware of the limitations of the assays. Of course, several open questions are still under research and because of it cfDNA and ctDNA testing are not part of routine care yet.
Collapse
Affiliation(s)
- András Telekes
- Omnimed-Etosz, Ltd., 81 Széher Rd., 1021 Budapest, Hungary
- Semmelweis University, 26. Üllői Rd., 1085 Budapest, Hungary
| | - Anna Horváth
- Department of Internal Medicine and Haematology, Semmelweis University, 46. Szentkirályi Rd., 1088 Budapest, Hungary
| |
Collapse
|
55
|
Hsiehchen D, Bucheit L, Yang D, Beg MS, Lim M, Lee SS, Kasi PM, Kaseb AO, Zhu H. Genetic features and therapeutic relevance of emergent circulating tumor DNA alterations in refractory non-colorectal gastrointestinal cancers. Nat Commun 2022; 13:7477. [PMID: 36463294 PMCID: PMC9719461 DOI: 10.1038/s41467-022-35144-1] [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] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Acquired resistance to systemic treatments is inevitable in most cancers, but the genetic basis for this in many cancer types has remained elusive due to constraints in obtaining tissue specimens longitudinally. In the management of gastrointestinal cancers, molecular profiling is conventionally performed at a single time point, although serial evaluations may yield biological insights that inform treatment decisions. We characterize genetic changes in serial liquid biopsies which provide real-time snapshots of tumor genetics and heterogeneity in refractory non-colorectal gastrointestinal cancers, and determine the clinical utility of repeat circulating tumor DNA (ctDNA) testing. In a national cohort of 449 patients with pancreatic, biliary, esophagogastric, and hepatocellular cancers, resistance to conventional therapies is broadly associated with tumor evolution. Emergent ctDNA alterations only detectable at progression occurs in 63% of patients and are frequently associated with treatment actionability. Tumor mutation burden is dynamic in cancers undergoing treatment, but is not associated with time to progression. Objective tumor responses in a case series of patients receiving treatment matched to emergent alterations show that repeat liquid biopsies may have clinical benefit by expanding treatment options in advanced gastrointestinal cancers.
Collapse
Affiliation(s)
- David Hsiehchen
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | | - Dong Yang
- Guardant Health Inc, Redwood City, CA, USA
| | - Muhammad Shaalan Beg
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mir Lim
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sunyoung S Lee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pashtoon Murtaza Kasi
- Weill Cornell Medicine, Englander Institute of Precision Medicine, Meyer Cancer Center, New York, NY, USA
| | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hao Zhu
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
56
|
Mauri G, Patelli G, Gori V, Lauricella C, Mussolin B, Amatu A, Bencardino K, Tosi F, Bonazzina E, Bonoldi E, Bardelli A, Siena S, Sartore-Bianchi A. Case Report: MAP2K1 K57N mutation is associated with primary resistance to anti-EGFR monoclonal antibodies in metastatic colorectal cancer. Front Oncol 2022; 12:1030232. [PMID: 36419886 PMCID: PMC9676472 DOI: 10.3389/fonc.2022.1030232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
BackgroundWe aim to identify the prevalence and the role of the MAP2K1 K57N mutation in predicting resistance to anti-EGFR agents in metastatic colorectal cancer (mCRC) patients.MethodsWe retrospectively reviewed tumor-based next generation sequencing (NGS) results from mCRC patients screened for enrollment in the GO40872/STARTRK-2 clinical trial between July 2019 and March 2021. Then, in patients harboring microsatellite stable (MSS) RAS and BRAF wild-type MAP2K1 mutant mCRC, we reviewed outcome to treatment with anti-EGFR monoclonal antibodies.ResultsA total of 246 mCRC patients were screened. Most of them, 215/220 (97.7%), were diagnosed with MSS mCRC and 112/215 (52.1%) with MSS, RAS and BRAF wild-type mCRC. Among the latter, 2/112 (1.8%) had MAP2K1 K57N mutant mCRC and both received anti-EGFR monotherapy as third line treatment. In both patients, MAP2K1 K57N mutant tumors proved primary resistant to anti-EGFR agent panitumumab monotherapy. Of interest, one of these patients was treated with anti-EGFR agents three times throughout his course of treatment, achieving some clinical benefit only when associated with other cytotoxic agents (FOLFOX or irinotecan).ConclusionWe verified in a clinical real-world setting that MAP2K1 K57N mutation is a resistance mechanism to anti-EGFR agents in mCRC. Thus, we suggest avoiding the administration of these drugs to MSS RAS and BRAF wild-type MAP2K1 N57K mutant mCRC.
Collapse
Affiliation(s)
- Gianluca Mauri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- IFOM, Istituto Fondazione di Oncologia Molecolare ETS, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Viviana Gori
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Calogero Lauricella
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Emanuela Bonoldi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
- Department of Oncology, Università degli Studi di Torino, Turin, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- *Correspondence: Andrea Sartore-Bianchi,
| |
Collapse
|
57
|
Zhuo Z, Lin L, Miao L, Li M, He J. Advances in liquid biopsy in neuroblastoma. FUNDAMENTAL RESEARCH 2022; 2:903-917. [PMID: 38933377 PMCID: PMC11197818 DOI: 10.1016/j.fmre.2022.08.005] [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: 04/27/2022] [Revised: 07/18/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022] Open
Abstract
Even with intensive treatment of high-risk neuroblastoma (NB) patients, half of high-risk NB patients still relapse. New therapies targeting the biological characteristics of NB have important clinical value for the personalized treatment of NB. However, the current biological markers for NB are mainly analyzed by tissue biopsy. In recent years, circulating biomarkers of NB based on liquid biopsy have attracted more and more attention. This review summarizes the analytes and methods for liquid biopsy of NB. We focus on the application of liquid biopsy in the diagnosis, prognosis assessment, and monitoring of NB. Finally, we discuss the prospects and challenges of liquid biopsy in NB.
Collapse
Affiliation(s)
- Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
- Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Lei Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
58
|
Bożyk A, Nicoś M. The Overview of Perspectives of Clinical Application of Liquid Biopsy in Non-Small-Cell Lung Cancer. Life (Basel) 2022; 12:1640. [PMID: 36295075 PMCID: PMC9604747 DOI: 10.3390/life12101640] [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: 09/20/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 01/19/2023] Open
Abstract
The standard diagnostics procedure for non-small-cell lung cancer (NSCLC) requires a pathological evaluation of tissue samples obtained by surgery or biopsy, which are considered invasive sampling procedures. Due to this fact, re-sampling of the primary tumor at the moment of progression is limited and depends on the patient's condition, even if it could reveal a mechanism of resistance to applied therapy. Recently, many studies have indicated that liquid biopsy could be provided for the noninvasive management of NSCLC patients who receive molecularly targeted therapies or immunotherapy. The liquid biopsy of neoplastic patients harbors small fragments of circulating-free DNA (cfDNA) and cell-free RNA (cfRNA) secreted to the circulation from normal cells, as well as a subset of tumor-derived circulating tumor cells (CTCs) or circulating tumor DNA (ctDNA). In NSCLC patients, a longitudinal assessment of genetic alterations in "druggable" genes in liquid biopsy might improve the follow-up of treatment efficacy and allow for the detection of an early progression before it is detectable in computed tomography or a clinical image. However, a liquid biopsy may be used to determine a variety of relevant molecular or genetic information for understanding tumor biology and its evolutionary trajectories. Thus, liquid biopsy is currently associated with greater hope for common diagnostic and clinical applications. In this review, we would like to highlight diagnostic challenges in the application of liquid biopsy into the clinical routine and indicate its implications on the metastatic spread of NSCLC or monitoring of personalized treatment regimens.
Collapse
Affiliation(s)
| | - Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
59
|
Investigation of radiomics based intra-patient inter-tumor heterogeneity and the impact of tumor subsampling strategies. Sci Rep 2022; 12:17244. [PMID: 36241749 PMCID: PMC9568579 DOI: 10.1038/s41598-022-20931-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/21/2022] [Indexed: 01/06/2023] Open
Abstract
While radiomics analysis has been applied for localized cancer disease, its application to the metastatic setting involves a non-exhaustive lesion subsampling strategy which may sidestep the intrapatient tumoral heterogeneity, hindering the reproducibility and the therapeutic response performance. Our aim was to evaluate if radiomics features can capture intertumoral intrapatient heterogeneity, and the impact of tumor subsampling on the computed heterogeneity. To this end, We delineated and extracted radiomics features of all visible tumors from single acquisition pre-treatment computed tomography of patients with metastatic lung cancer (cohort L) and confirmed our results on a larger cohort of patients with metastatic melanoma (cohort M). To quantify the captured heterogeneity, the absolute coefficient of variation (CV) of each radiomics index was calculated at the patient-level and a sensitivity analysis was performed using only a subset of all extracted features robust to the segmentation step. The extent of information loss by six commonly used tumor sampling strategies was then assessed. A total of 602 lesions were segmented from 43 patients (median age 57, 4.9% female). All robust radiomics indexes exhibited at least 20% of variation with significant heterogeneity both in heavily and oligo metastasized patients, and also at the organ level. None of the segmentation subsampling strategies were able to recover the true tumoral heterogeneity obtained by exhaustive tumor sampling. Image-based inter-tumor intra-patient heterogeneity can be successfully grasped by radiomics analyses. Failing to take into account this kind of heterogeneity will lead to inconsistent predictive algorithms. Guidelines to standardize the tumor sampling step and/or AI-driven tools to alleviate the segmentation effort are required.
Collapse
|
60
|
Bai X, Quek C. Unravelling Tumour Microenvironment in Melanoma at Single-Cell Level and Challenges to Checkpoint Immunotherapy. Genes (Basel) 2022; 13:genes13101757. [PMID: 36292642 PMCID: PMC9601741 DOI: 10.3390/genes13101757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Melanoma is known as one of the most immunogenic tumours and is often characterised by high mutation burden, neoantigen load and immune infiltrate. The application of immunotherapies has led to impressive improvements in the clinical outcomes of advanced stage melanoma patients. The standard of care immunotherapies leverage the host immunological influence on tumour cells, which entail complex interactions among the tumour, stroma, and immune cells at the tumour microenvironmental level. However, not all cancer patients can achieve a long-term durable response to immunotherapy, and a significant proportion of patients develops resistance and still die from their disease. Owing to the multi-faceted problems of tumour and microenvironmental heterogeneity, identifying the key factors underlying tumour progression and immunotherapy resistance poses a great challenge. In this review, we outline the main challenges to current cancer immunotherapy research posed by tumour heterogeneity and microenvironment complexities including genomic and transcriptomic variability, selective outgrowth of tumour subpopulations, spatial and temporal tumour heterogeneity and the dynamic state of host immunity and microenvironment orchestration. We also highlight the opportunities to dissect tumour heterogeneity using single-cell sequencing and spatial platforms. Integrative analyses of large-scale datasets will enable in-depth exploration of biological questions, which facilitates the clinical application of translational research.
Collapse
|
61
|
Vitkin E, Singh A, Wise J, Ben-Elazar S, Yakhini Z, Golberg A. Nondestructive protein sampling with electroporation facilitates profiling of spatial differential protein expression in breast tumors in vivo. Sci Rep 2022; 12:15835. [PMID: 36151122 PMCID: PMC9508265 DOI: 10.1038/s41598-022-19984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
Excision tissue biopsy, while central to cancer treatment and precision medicine, presents risks to the patient and does not provide a sufficiently broad and faithful representation of the heterogeneity of solid tumors. Here we introduce e-biopsy—a novel concept for molecular profiling of solid tumors using molecular sampling with electroporation. As e-biopsy provides access to the molecular composition of a solid tumor by permeabilization of the cell membrane, it facilitates tumor diagnostics without tissue resection. Furthermore, thanks to its non tissue destructive characteristics, e-biopsy enables probing the solid tumor multiple times in several distinct locations in the same procedure, thereby enabling the spatial profiling of tumor molecular heterogeneity.We demonstrate e-biopsy in vivo, using the 4T1 breast cancer model in mice to assess its performance, as well as the inferred spatial differential protein expression. In particular, we show that proteomic profiles obtained via e-biopsy in vivo distinguish the tumors from healthy breast tissue and reflect spatial tumor differential protein expression. E-biopsy provides a completely new molecular sampling modality for solid tumors molecular cartography, providing information that potentially enables more rapid and sensitive detection at lesser risk, as well as more precise personalized medicine.
Collapse
Affiliation(s)
- Edward Vitkin
- School of Computer Science, Reichman University (IDC Herzliya), Herzliya, Israel
| | - Amrita Singh
- Porter School of Environment and Earth Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Julia Wise
- Porter School of Environment and Earth Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shay Ben-Elazar
- School of Computer Science, Reichman University (IDC Herzliya), Herzliya, Israel
| | - Zohar Yakhini
- School of Computer Science, Reichman University (IDC Herzliya), Herzliya, Israel. .,Computer Science Faculty, Technion, Haifa, Israel.
| | - Alexander Golberg
- Porter School of Environment and Earth Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
62
|
Hewitt DB, Brown ZJ, Pawlik TM. The Role of Biomarkers in the Management of Colorectal Liver Metastases. Cancers (Basel) 2022; 14:cancers14194602. [PMID: 36230522 PMCID: PMC9559307 DOI: 10.3390/cancers14194602] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Colorectal cancer remains one of the most significant sources of cancer-related morbidity and mortality worldwide. The liver is the most common site of metastatic spread. Multiple modalities exist to manage and potentially cure patients with metastatic colorectal cancer. However, reliable biomarkers to assist with clinical decision-making are limited. Recent advances in genomic sequencing technology have greatly expanded our knowledge of colorectal cancer carcinogenesis and significantly reduced the cost and timing of the investigation. In this article, we discuss the current utility of biomarkers in the management of colorectal cancer liver metastases. Abstract Surgical management combined with improved systemic therapies have extended 5-year overall survival beyond 50% among patients with colorectal liver metastases (CRLM). Furthermore, a multitude of liver-directed therapies has improved local disease control for patients with unresectable CRLM. Unfortunately, a significant portion of patients treated with curative-intent hepatectomy develops disease recurrence. Traditional markers fail to risk-stratify and prognosticate patients with CRLM appropriately. Over the last few decades, advances in molecular sequencing technology have greatly expanded our knowledge of the pathophysiology and tumor microenvironment characteristics of CRLM. These investigations have revealed biomarkers with the potential to better inform management decisions in patients with CRLM. Actionable biomarkers such as RAS and BRAF mutations, microsatellite instability/mismatch repair status, and tumor mutational burden have been incorporated into national and societal guidelines. Other biomarkers, including circulating tumor DNA and radiomic features, are under active investigation to evaluate their clinical utility. Given the plethora of therapeutic modalities and lack of evidence on timing and sequence, reliable biomarkers are needed to assist clinicians with the development of patient-tailored management plans. In this review, we discuss the current evidence regarding biomarkers for patients with CRLM.
Collapse
|
63
|
Synthesis of Silver Nano Particles Using Myricetin and the In-Vitro Assessment of Anti-Colorectal Cancer Activity: In-Silico Integration. Int J Mol Sci 2022; 23:ijms231911024. [PMID: 36232319 PMCID: PMC9570303 DOI: 10.3390/ijms231911024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/29/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
The creation of novel anticancer treatments for a variety of human illnesses, including different malignancies and dangerous microbes, also potentially depends on nanoparticles including silver. Recently, it has been successful to biologically synthesize metal nanoparticles using plant extracts. The natural flavonoid 3,3′, 4′, 5,5′, and 7 hexahydroxyflavon (myricetin) has anticancer properties. There is not much known about the regulatory effects of myricetin on the possible cell fate-determination mechanisms (such as apoptosis/proliferation) in colorectal cancer. Because the majority of investigations related to the anticancer activity of myricetin have dominantly focused on the enhancement of tumor cell uncontrolled growth (i.e., apoptosis). Thus, we have decided to explore the potential myricetin interactors and the associated biological functions by using an in-silico approach. Then, we focused on the main goal of the work which involved the synthesis of silver nanoparticles and the labeling of myricetin with it. The synthesized silver nanoparticles were examined using UV-visible spectroscopy, dynamic light scattering spectroscopy, Fourier transform infrared spectroscopy, and scanning electron microscopy. In this study, we have investigated the effects of myricetin on colorectal cancer where numerous techniques were used to show myricetin’s effect on colon cancer cells. Transmission Electron Microscopy was employed to monitor morphological changes. Furthermore, we have combined the results of the colorectal cancer gene expression dataset with those of the myricetin interactors and pathways. Based on the results, we conclude that myricetin is able to efficiently kill human colorectal cancer cell lines. Since, it shares important biological roles and possible route components and this myricetin may be a promising herbal treatment for colorectal cancer as per an in-silico analysis of the TCGA dataset.
Collapse
|
64
|
Johnson RM, Qu X, Lin CF, Huw LY, Venkatanarayan A, Sokol E, Ou FS, Ihuegbu N, Zill OA, Kabbarah O, Wang L, Bourgon R, de Sousa E Melo F, Bolen C, Daemen A, Venook AP, Innocenti F, Lenz HJ, Bais C. ARID1A mutations confer intrinsic and acquired resistance to cetuximab treatment in colorectal cancer. Nat Commun 2022; 13:5478. [PMID: 36117191 PMCID: PMC9482920 DOI: 10.1038/s41467-022-33172-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
Most colorectal (CRC) tumors are dependent on EGFR/KRAS/BRAF/MAPK signaling activation. ARID1A is an epigenetic regulator mutated in approximately 5% of non-hypermutated CRC tumors. Here we show that anti-EGFR but not anti-VEGF treatment enriches for emerging ARID1A mutations in CRC patients. In addition, we find that patients with ARID1A mutations, at baseline, are associated with worse outcome when treated with cetuximab- but not bevacizumab-containing therapies; thus, this suggests that ARID1A mutations may provide both an acquired and intrinsic mechanism of resistance to anti-EGFR therapies. We find that, ARID1A and EGFR-pathway genetic alterations are mutually exclusive across lung and colorectal cancers, further supporting a functional connection between these pathways. Our results not only suggest that ARID1A could be potentially used as a predictive biomarker for cetuximab treatment decisions but also provide a rationale for exploring therapeutic MAPK inhibition in an unexpected but genetically defined segment of CRC patients. ARID1A is an epigenetic regulator mutated in approximately 5% of non-hypermutated colorectal cancer tumors, however, its relationship with treatment response remains to be explored. Here, the authors suggest that ARID1A mutations may confer intrinsic and acquired resistance to cetuximab treatment.
Collapse
Affiliation(s)
- Radia M Johnson
- Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA, USA.
| | - Xueping Qu
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA.
| | - Chu-Fang Lin
- Real World Data Science Analytics, Genentech, Inc., South San Francisco, CA, USA
| | - Ling-Yuh Huw
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | | | - Ethan Sokol
- Cancer Genomics Research, Foundation Medicine, Inc., Cambridge, MA, USA
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | | | - Oliver A Zill
- Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA, USA
| | - Omar Kabbarah
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Lisa Wang
- Real World Data Science Analytics, Genentech, Inc., South San Francisco, CA, USA
| | - Richard Bourgon
- Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA, USA
| | | | - Chris Bolen
- Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA, USA
| | - Anneleen Daemen
- Bioinformatics & Computational Biology, Genentech, Inc., South San Francisco, CA, USA
| | - Alan P Venook
- University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Carlos Bais
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
65
|
Scheiter A, Hierl F, Winkel I, Keil F, Klier-Richter M, Coulouarn C, Lüke F, Kandulski A, Evert M, Dietmaier W, Calvisi DF, Utpatel K. Wnt/β-Catenin-Pathway Alterations and Homologous Recombination Deficiency in Cholangiocarcinoma Cell Lines and Clinical Samples: Towards Specific Vulnerabilities. J Pers Med 2022; 12:1270. [PMID: 36013219 PMCID: PMC9410222 DOI: 10.3390/jpm12081270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 12/01/2022] Open
Abstract
Cholangiocarcinoma (CCA) features a dismal prognosis with limited treatment options. Genomic studies have unveiled several promising targets in this disease, including fibroblast growth factor receptor (FGFR) fusions and isocitrate dehydrogenase (IDH) mutations. To fully harness the potential of genomically informed therapies in CCA, it is necessary to thoroughly characterize the available model organisms, including cell lines. One parameter to investigate in CCA is homologous recombination deficiency (HRD). While mutations in homologous recombinational repair (HRR)-related genes have been detected, their predictive value remains undetermined. Using a targeted next-generation sequencing approach, we analyzed 12 human CCA cell lines and compared them to 62 CCA samples of the molecular tumor board cohort. The AmoyDx® HRD Focus Panel was employed to determine corresponding genomic scar scores (GSS). Ten of twelve cell lines harbored alterations in common HRR-related genes, and five cell lines were HRD-positive, although this parameter did not correlate well with Olaparib sensitivity. Moreover, functionally relevant APC and β-catenin mutations were registered, which were also detected in 4/176 (2.3%) samples on a CCA microarray. Although rare, these alterations were exclusive to large duct type CCA with associated intraductal papillary neoplasms of the bile duct (IPNB) in 3 cases, pointing at a distinct form of cholangiocarcinogenesis with potential specific vulnerabilities.
Collapse
Affiliation(s)
- Alexander Scheiter
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
- Bavarian Center for Cancer Research/BZKF, 91054 Bavaria, Germany;
| | - Frederik Hierl
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | - Ingrid Winkel
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | - Felix Keil
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | - Margit Klier-Richter
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | | | - Florian Lüke
- Bavarian Center for Cancer Research/BZKF, 91054 Bavaria, Germany;
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93053 Regensburg, Germany
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, 93053 Regensburg, Germany
| | - Arne Kandulski
- Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | - Wolfgang Dietmaier
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | - Diego F. Calvisi
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| | - Kirsten Utpatel
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.H.); (I.W.); (F.K.); (M.K.-R.); (M.E.); (W.D.); (D.F.C.); (K.U.)
| |
Collapse
|
66
|
Mauri G, Vitiello PP, Sogari A, Crisafulli G, Sartore-Bianchi A, Marsoni S, Siena S, Bardelli A. Liquid biopsies to monitor and direct cancer treatment in colorectal cancer. Br J Cancer 2022; 127:394-407. [PMID: 35264786 PMCID: PMC9346106 DOI: 10.1038/s41416-022-01769-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/02/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and deadly cancers worldwide. Despite recent improvements in treatment and prevention, most of the current therapeutic options are weighted by side effects impacting patients' quality of life. Better patient selection towards systemic treatments represents an unmet clinical need. The recent multidisciplinary and molecular advancements in the treatment of CRC patients demand the identification of efficient biomarkers allowing to personalise patient care. Currently, core tumour biopsy specimens represent the gold-standard biological tissue to identify such biomarkers. However, technical feasibility, tumour heterogeneity and cancer evolution are major limitations of this single-snapshot approach. Genotyping circulating tumour DNA (ctDNA) has been addressed as potentially overcoming such limitations. Indeed, ctDNA has been retrospectively demonstrated capable of identifying minimal residual disease post-surgery and post-adjuvant treatment, as well as spotting druggable molecular alterations for tailoring treatments in metastatic disease. In this review, we summarise the available evidence on ctDNA applicability in CRC. Then, we review ongoing clinical trials assessing how liquid biopsy can be used interventionally to guide therapeutic choice in localised, locally advanced and metastatic CRC. Finally, we discuss how its widespread could transform CRC patients' management, dissecting its limitations while suggesting improvement strategies.
Collapse
Affiliation(s)
- Gianluca Mauri
- IFOM-FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Pietro Paolo Vitiello
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Torino, 10060, Candiolo, TO, Italy
| | - Alberto Sogari
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Torino, 10060, Candiolo, TO, Italy
| | - Giovanni Crisafulli
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Torino, 10060, Candiolo, TO, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | | | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy.
- Department of Oncology, University of Torino, 10060, Candiolo, TO, Italy.
| |
Collapse
|
67
|
Sartore-Bianchi A, Pietrantonio F, Lonardi S, Mussolin B, Rua F, Crisafulli G, Bartolini A, Fenocchio E, Amatu A, Manca P, Bergamo F, Tosi F, Mauri G, Ambrosini M, Daniel F, Torri V, Vanzulli A, Regge D, Cappello G, Marchiò C, Berrino E, Sapino A, Marsoni S, Siena S, Bardelli A. Circulating tumor DNA to guide rechallenge with panitumumab in metastatic colorectal cancer: the phase 2 CHRONOS trial. Nat Med 2022; 28:1612-1618. [PMID: 35915157 PMCID: PMC9386661 DOI: 10.1038/s41591-022-01886-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 06/01/2022] [Indexed: 01/09/2023]
Abstract
Anti-epidermal growth factor receptor (EGFR) monoclonal antibodies are approved for the treatment of RAS wild-type (WT) metastatic colorectal cancer (mCRC), but the emergence of resistance mutations restricts their efficacy. We previously showed that RAS, BRAF and EGFR mutant alleles, which appear in circulating tumor DNA (ctDNA) during EGFR blockade, decline upon therapy withdrawal. We hypothesized that monitoring resistance mutations in blood could rationally guide subsequent therapy with anti-EGFR antibodies. We report here the results of CHRONOS, an open-label, single-arm phase 2 clinical trial exploiting blood-based identification of RAS/BRAF/EGFR mutations levels to tailor a chemotherapy-free anti-EGFR rechallenge with panitumumab (ClinicalTrials.gov: NCT03227926 ; EudraCT 2016-002597-12). The primary endpoint was objective response rate. Secondary endpoints were progression-free survival, overall survival, safety and tolerability of this strategy. In CHRONOS, patients with tissue-RAS WT tumors after a previous treatment with anti-EGFR-based regimens underwent an interventional ctDNA-based screening. Of 52 patients, 16 (31%) carried at least one mutation conferring resistance to anti-EGFR therapy and were excluded. The primary endpoint of the trial was met; and, of 27 enrolled patients, eight (30%) achieved partial response and 17 (63%) disease control, including two unconfirmed responses. These clinical results favorably compare with standard third-line treatments and show that interventional liquid biopsies can be effectively and safely exploited in a timely manner to guide anti-EGFR rechallenge therapy with panitumumab in patients with mCRC. Further larger and randomized trials are warranted to formally compare panitumumab rechallenge with standard-of-care therapies in this patient setting.
Collapse
Affiliation(s)
- Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Medical Oncology Unit 1, Veneto Institute of Oncology IOV-IRCCS Padua, Padua, Italy
| | | | | | - Giovanni Crisafulli
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Oncology, University of Turin, Turin, Italy
| | | | | | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Paolo Manca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Bergamo
- Medical Oncology Unit 1, Veneto Institute of Oncology IOV-IRCCS Padua, Padua, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Gianluca Mauri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
- IFOM, FIRC Institute of Molecular Oncology, Milan, Italy
| | - Margherita Ambrosini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Daniel
- Medical Oncology Unit 1, Veneto Institute of Oncology IOV-IRCCS Padua, Padua, Italy
| | - Valter Torri
- Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy
| | - Angelo Vanzulli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
- Department of Services, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Daniele Regge
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Unit of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Giovanni Cappello
- Unit of Radiology, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Enrico Berrino
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Marsoni
- IFOM, FIRC Institute of Molecular Oncology, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy.
- Department of Oncology, University of Turin, Turin, Italy.
| |
Collapse
|
68
|
Vaquero J, Pavy A, Gonzalez-Sanchez E, Meredith M, Arbelaiz A, Fouassier L. Genetic alterations shaping tumor response to anti-EGFR therapies. Drug Resist Updat 2022; 64:100863. [DOI: 10.1016/j.drup.2022.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
69
|
Russo M, Pompei S, Sogari A, Corigliano M, Crisafulli G, Puliafito A, Lamba S, Erriquez J, Bertotti A, Gherardi M, Di Nicolantonio F, Bardelli A, Cosentino Lagomarsino M. A modified fluctuation-test framework characterizes the population dynamics and mutation rate of colorectal cancer persister cells. Nat Genet 2022; 54:976-984. [PMID: 35817983 PMCID: PMC9279152 DOI: 10.1038/s41588-022-01105-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/25/2022] [Indexed: 12/16/2022]
Abstract
Compelling evidence shows that cancer persister cells represent a major limit to the long-term efficacy of targeted therapies. However, the phenotype and population dynamics of cancer persister cells remain unclear. We developed a quantitative framework to study persisters by combining experimental characterization and mathematical modeling. We found that, in colorectal cancer, a fraction of persisters slowly replicates. Clinically approved targeted therapies induce a switch to drug-tolerant persisters and a temporary 7- to 50-fold increase of their mutation rate, thus increasing the number of persister-derived resistant cells. These findings reveal that treatment may influence persistence and mutability in cancer cells and pinpoint inhibition of error-prone DNA polymerases as a strategy to restrict tumor recurrence.
Collapse
Affiliation(s)
- Mariangela Russo
- Department of Oncology, University of Turin, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Simone Pompei
- IFOM Foundation, FIRC Institute of Molecular Oncology, Milan, Italy
| | - Alberto Sogari
- Department of Oncology, University of Turin, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Mattia Corigliano
- IFOM Foundation, FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Physics, University of Milan and INFN, Milan, Italy
| | - Giovanni Crisafulli
- Department of Oncology, University of Turin, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Alberto Puliafito
- Department of Oncology, University of Turin, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Simona Lamba
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Andrea Bertotti
- Department of Oncology, University of Turin, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Marco Gherardi
- IFOM Foundation, FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Physics, University of Milan and INFN, Milan, Italy
| | - Federica Di Nicolantonio
- Department of Oncology, University of Turin, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Alberto Bardelli
- Department of Oncology, University of Turin, Candiolo, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| | - Marco Cosentino Lagomarsino
- IFOM Foundation, FIRC Institute of Molecular Oncology, Milan, Italy.
- Department of Physics, University of Milan and INFN, Milan, Italy.
| |
Collapse
|
70
|
Wu W, Li X, Yu S. Patient-derived Tumour Organoids: A Bridge between Cancer Biology and Personalised Therapy. Acta Biomater 2022; 146:23-36. [PMID: 35533925 DOI: 10.1016/j.actbio.2022.04.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 02/06/2023]
Abstract
Patient-derived tumour organoids (PDOs) have revolutionised our understanding of cancer biology and the applications of personalised therapies. These advancements are principally ascribed to the ability of PDOs to consistently recapitulate and maintain the genomic, proteomic and morphological characteristics of parental tumours. Given these characteristics, PDOs (and their extended biobanks) are a representative preclinical model eminently suited to translate relevant scientific findings into personalized therapies rapidly. Here, we summarise recent advancements in PDOs from the perspective of cancer biology and clinical applications, focusing on the current challenges and opportunities of reconstructing and standardising more sophisticated PDO models. STATEMENT OF SIGNIFICANCE: Patient-derived tumour organoids (PDOs), three-dimensional (3D) self-assembled organotypic structures, have revolutionised our understanding of cancer biology and the applications of personalised therapies. These advancements are principally ascribed to the ability of PDOs to consistently recapitulate and maintain the genomic, proteomic and morphological characteristics of parental tumours. Given these characteristics, PDOs (and their extended biobanks) are a representative preclinical model eminently suited to translate relevant scientific findings into personalized therapies rapidly.
Collapse
Affiliation(s)
- Wence Wu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Xiaoyang Li
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Shengji Yu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
71
|
Ceccon C, Angerilli V, Rasola C, Procaccio L, Sabbadin M, Bergamo F, Malapelle U, Lonardi S, Fassan M. Microsatellite Instable Colorectal Adenocarcinoma Diagnostics: The Advent of Liquid Biopsy Approaches. Front Oncol 2022; 12:930108. [PMID: 35837109 PMCID: PMC9273960 DOI: 10.3389/fonc.2022.930108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/30/2022] [Indexed: 11/24/2022] Open
Abstract
The introduction of immunotherapy has revolutionized the oncological targeted therapy paradigm. Microsatellite instability (MSI) identifies a subgroup of colorectal cancers (CRCs) which respond to treatment with immune checkpoint inhibitors. Tissue biopsy is currently the gold standard for the assessment of MSI/Mismatch Repair deficiency (MMRd) by means immunohistochemistry or molecular assays. However, the application of liquid biopsy in the clinic may help to overcome several limitations of tissue analysis and may provide great benefit to the diagnostic scenario and therapeutic decision-making process. In the context of MSI/MMRd CRC, the use of liquid biopsy may allow to establish MSI/MMR status if tissue sampling cannot be performed or in case of discordant tissue biopsies. Liquid biopsy may also become a powerful tool to monitor treatment response and the onset resistance to immunotherapy over time and to stratify of MSI/MMRd patients according to their risk of relapse and metastases. The aim of this review is to summarize the main technical aspects and clinical applications, the benefits, and limitations of the use of liquid biopsy in MSI/MMRd colorectal cancer patients.
Collapse
Affiliation(s)
- Carlotta Ceccon
- Department of Medicine (DIMED), University of Padua, Padua, Italy
| | | | - Cosimo Rasola
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | | | | | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Sara Lonardi
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| |
Collapse
|
72
|
Blood-based liquid biopsies for prostate cancer: clinical opportunities and challenges. Br J Cancer 2022; 127:1394-1402. [PMID: 35715640 PMCID: PMC9553885 DOI: 10.1038/s41416-022-01881-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 12/19/2022] Open
Abstract
Liquid biopsy has been established as a powerful, minimally invasive, tool to detect clinically actionable aberrations across numerous cancer types in real-time. With the development of new therapeutic agents in prostate cancer (PC) including DNA repair targeted therapies, this is especially attractive. However, there is unclarity on how best to screen for PC, improve risk stratification and ultimately how to treat advanced disease. Therefore, there is an urgent need to develop better biomarkers to help guide oncologists' decisions in these settings. Circulating tumour cells (CTCs), exosomes and cell-free DNA/RNA (cfDNA/cfRNA) analysis, including epigenetic features such as methylation, have all shown potential in prognostication, treatment response assessment and detection of emerging mechanisms of resistance. However, there are still challenges to overcome prior to implementing liquid biopsies in routine clinical practice such as preanalytical considerations including blood collection and storage, the cost of CTC isolation and enrichment, low-circulating tumour content as a limitation for genomic analysis and how to better interpret the sequencing data generated. In this review, we describe an overview of the up-to-date clinical opportunities in the management of PC through blood-based liquid biopsies and the next steps for its implementation in personalised treatment guidance.
Collapse
|
73
|
Fischer LE, Stintzing S, Heinemann V, Keilholz U, Keune D, Vollbrecht C, Burmeister T, Kind A, Weiss L, Horst D, Kirchner T, Klauschen F, Jung A, Westphalen CB, Jelas I. Liquid Biopsy in Colorectal Cancer: Quo Vadis? Implementation of Liquid Biopsies in Routine Clinical Patient Care in Two German Comprehensive Cancer Centers. Front Oncol 2022; 12:870411. [PMID: 35646657 PMCID: PMC9134071 DOI: 10.3389/fonc.2022.870411] [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: 02/06/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives The use of liquid biopsies (LB) in patients with solid malignancies enables comprehensive genomic profiling (CGP) of circulating tumor DNA (ctDNA) and has the potential to guide therapy stratification and support disease monitoring. To examine clinical uptake of LB in a real-world setting, LB implementation was analyzed at two German cancer centers (LMU Munich and Charité - Universitätsmedizin Berlin) between 2017 and 2021, with focus on colorectal cancer (CRC) patients. Methods In this retrospective analysis, all patients who received a LB between January 2017 and December 2021 as part of routine clinical management were included. To provide adequate context, we collected disease characteristics and technical specifications of the LB methods applied. Additionally, we examined the concordance of RAS status in tumor tissue and LB. Finally, we discuss the potential of LB as a diagnostic tool to drive personalized treatment in CRC patients and how to implement LB in clinical routine. Results In total, our cohort included 86 CRC patients and 161 LB conducted in these patients between 2017 and 2021. In 59 patients, comparison between tissue-based and liquid-based molecular diagnostics, revealed a divergence in 23 (39%) of the evaluable samples. Conclusion Our real-world data analysis indicates that the possibilities of LB are not yet exploited in everyday clinical practice. Currently, the variety of methods and lack of standardization, as well as restricted reimbursement for liquid based CGP hinder the use of LB in clinical routine. To overcome these issues, prospective clinical trials are needed to provide evidence driving the implementation of LB into the management of CRC patients and to support their implementation into clinical guidelines.
Collapse
Affiliation(s)
- Laura E Fischer
- Department of Medicine III, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Ulrich Keilholz
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany.,Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dietmar Keune
- Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudia Vollbrecht
- Institute of Pathology Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Burmeister
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Labor Berlin - Charité Vivantes, GmbH, Molekulardiagnostik - Hämatologie, Berlin, Germany
| | - Andreas Kind
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lena Weiss
- Department of Medicine III, University Hospital, Munich, Germany
| | - David Horst
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany.,Institute of Pathology Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Kirchner
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.,Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, München, Germany
| | - Frederick Klauschen
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.,Institute of Pathology Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, München, Germany
| | - Andreas Jung
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.,Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, München, Germany
| | - Christoph Benedikt Westphalen
- Department of Medicine III, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Ivan Jelas
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany
| |
Collapse
|
74
|
Liu Q, Li X, Chen H. Genomic evolution of cancer metastasis under therapeutic pressure. MEDCOMM – ONCOLOGY 2022. [DOI: 10.1002/mog2.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Qiu‐Luo Liu
- Department of Gastrointestinal Surgery, West China Hospital Sichuan University Chengdu Sichuan PR China
| | - Xi Li
- Colorectal Surgery Unit, Department of General Surgery, Addenbrooke's Hospital Cambridge University Hospitals NHS Foundation Trust Cambridge UK
| | - Hai‐Ning Chen
- Department of Gastrointestinal Surgery, West China Hospital Sichuan University Chengdu Sichuan PR China
| |
Collapse
|
75
|
Kim S, Lim Y, Kang JK, Kim HP, Roh H, Kim SY, Lee D, Bang D, Jeong SY, Park KJ, Han SW, Kim TY. Dynamic changes in longitudinal circulating tumour DNA profile during metastatic colorectal cancer treatment. Br J Cancer 2022; 127:898-907. [DOI: 10.1038/s41416-022-01837-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/09/2022] Open
|
76
|
Basic Science with Preclinical Models to Investigate and Develop Liquid Biopsy: What Are the Available Data and Is It a Fruitful Approach? Int J Mol Sci 2022; 23:ijms23105343. [PMID: 35628154 PMCID: PMC9141279 DOI: 10.3390/ijms23105343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
The molecular analysis of circulating analytes (circulating tumor-DNA (ctDNA), -cells (CTCs) and -RNA (ctRNA)/exosomes) deriving from solid tumors and detected in the bloodstream—referred as liquid biopsy—has emerged as one of the most promising concepts in cancer management. Compelling data have evidenced its pivotal contribution and unique polyvalence through multiple applications. These data essentially derived from translational research. Therewith, data on liquid biopsy in basic research with preclinical models are scarce, a concerning lack that has been widely acknowledged in the field. This report aimed to comprehensively review the available data on the topic, for each analyte. Only 17, 17 and 2 studies in basic research investigated ctDNA, CTCs and ctRNA/exosomes, respectively. Albeit rare, these studies displayed noteworthy relevance, demonstrating the capacity to investigate questions related to the biology underlying analytes release that could not be explored via translational research with human samples. Translational, clinical and technological sectors of liquid biopsy may benefit from basic research and should take note of some important findings generated by these studies. Overall, results underscored the need to intensify the efforts to conduct future studies on liquid biopsy in basic research with new preclinical models.
Collapse
|
77
|
Yaung SJ, Ju C, Gattam S, Nicholas A, Sommer N, Bendell JC, Hurwitz HI, Lee JJ, Casey F, Price R, Palma JF. Plasma-Based Measurements of Tumor Heterogeneity Correlate with Clinical Outcomes in Metastatic Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14092240. [PMID: 35565368 PMCID: PMC9105064 DOI: 10.3390/cancers14092240] [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: 03/18/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Sequencing circulating tumor DNA (ctDNA) from liquid biopsies may better assess tumor heterogeneity than limited sampling of tumor tissue. Here, we explore ctDNA-based heterogeneity and its correlation with treatment outcome in STEAM, which assessed efficacy and safety of concurrent and sequential FOLFOXIRI-bevacizumab (BEV) vs. FOLFOX-BEV for first-line treatment of metastatic colorectal cancer. We sequenced 146 pre-induction and 89 post-induction patient plasmas with a 198-kilobase capture-based assay, and applied Mutant-Allele Tumor Heterogeneity (MATH), a traditionally tissue-based calculation of allele frequency distribution, on somatic mutations detected in plasma. Higher levels of MATH, particularly in the post-induction sample, were associated with shorter progression-free survival (PFS). Patients with high MATH vs. low MATH in post-induction plasma had shorter PFS (7.2 vs. 11.7 months; hazard ratio, 3.23; 95% confidence interval, 1.85−5.63; log-rank p < 0.0001). These results suggest ctDNA-based tumor heterogeneity may have potential prognostic value in metastatic cancers.
Collapse
Affiliation(s)
- Stephanie J. Yaung
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (J.J.L.); (F.C.); (J.F.P.)
- Correspondence: ; Tel.: +1-925-523-8824
| | - Christine Ju
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.)
| | - Sandeep Gattam
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.)
| | - Alan Nicholas
- Genentech, Inc., South San Francisco, CA 94080, USA; (A.N.); (N.S.); (H.I.H.); (R.P.)
| | - Nicolas Sommer
- Genentech, Inc., South San Francisco, CA 94080, USA; (A.N.); (N.S.); (H.I.H.); (R.P.)
| | - Johanna C. Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN 37203, USA;
| | - Herbert I. Hurwitz
- Genentech, Inc., South San Francisco, CA 94080, USA; (A.N.); (N.S.); (H.I.H.); (R.P.)
| | - John J. Lee
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (J.J.L.); (F.C.); (J.F.P.)
| | - Fergal Casey
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (J.J.L.); (F.C.); (J.F.P.)
| | - Richard Price
- Genentech, Inc., South San Francisco, CA 94080, USA; (A.N.); (N.S.); (H.I.H.); (R.P.)
| | - John F. Palma
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (J.J.L.); (F.C.); (J.F.P.)
| |
Collapse
|
78
|
Ye JJ, Yu W, Xie BR, Li K, Liu MD, Dong X, Chen ZX, Feng J, Zhang XZ. Self-Reinforced Cancer Targeting (SRCT) Depending on Reciprocally Enhancing Feedback between Targeting and Therapy. ACS NANO 2022; 16:5851-5866. [PMID: 35412799 DOI: 10.1021/acsnano.1c10999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Conventional cancer targeting methodology needs to be reformed to overcome the intrinsic barriers responsible for poor targeting efficiency. This study describes a concept of self-reinforced cancer targeting (SRCT) by correlating targeting with therapy in a reciprocally enhancing manner. SRCT is achieved on the basis of two prerequisites: (1) target molecules have to be expressed on cancer cell membranes but not on normal cells, and (2) notably, their expression on cancer cells must be actively upregulated in response to cellular attack by cancer treatments. As a proof-of-concept, a GRP78-targeting nanovehicle for chemotherapy was designed. Resultant data showed that chemotherapeutic drugs could effectively elevate GRP78 expression on the plasma membranes of cancer cells while having minimal influence on normal cells. DOX pretreatment of cancer cells and tumor tissues can greatly increase the targeting efficacy and therapeutic performance of the prepared GRP78-targeting nanomedicine while somewhat disfavoring the nontargeting counterpart. In vivo and in vitro results demonstrated that this GRP78-targeting nanomedicine could accurately target cancer cells to not only implement chemotherapy but also induce GRP78 upregulation on cancer cells, eventually benefiting continuous cancer-cell-targeted attack by the nanomedicines remaining in the blood circulation or administered in the next dose. The GRP78-targeting nanomedicine displays much better antitumor performance compared with the nontargeting counterpart. SRCT is expected to advance cancer-targeted therapy based on the positive dependency between targeting and therapeutic modalities.
Collapse
Affiliation(s)
- Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Wuyang Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Bo-Ru Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Ke Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Miao-Deng Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan 430072, P.R. China
| | - Zhao-Xia Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P.R. China
| |
Collapse
|
79
|
Jia Q, Wang A, Yuan Y, Zhu B, Long H. Heterogeneity of the tumor immune microenvironment and its clinical relevance. Exp Hematol Oncol 2022; 11:24. [PMID: 35461288 PMCID: PMC9034473 DOI: 10.1186/s40164-022-00277-y] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/10/2022] [Indexed: 02/08/2023] Open
Abstract
During the course of tumorigenesis and subsequent metastasis, malignant cells gradually diversify and become more heterogeneous. Consequently, the tumor mass might be infiltrated by diverse immune-related components, including the cytokine/chemokine environment, cytotoxic activity, or immunosuppressive elements. This immunological heterogeneity is universally presented spatially or varies temporally along with tumor evolution or therapeutic intervention across almost all solid tumors. The heterogeneity of anti-tumor immunity shows a profound association with the progression of disease and responsiveness to treatment, particularly in the realm of immunotherapy. Therefore, an accurate understanding of tumor immunological heterogeneity is essential for the development of effective therapies. Facilitated by multi-regional and -omics sequencing, single cell sequencing, and longitudinal liquid biopsy approaches, recent studies have demonstrated the potential to investigate the complexity of immunological heterogeneity of the tumors and its clinical relevance in immunotherapy. Here, we aimed to review the mechanism underlying the heterogeneity of the immune microenvironment. We also explored how clinical assessments of tumor heterogeneity might facilitate the development of more effective personalized therapies.
Collapse
Affiliation(s)
- Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Army Military Medical University, Xinqiao Main Street, Chongqing, 400037, China.,Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Aoyun Wang
- Institute of Cancer, Xinqiao Hospital, Army Military Medical University, Xinqiao Main Street, Chongqing, 400037, China.,Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yixiao Yuan
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Army Military Medical University, Xinqiao Main Street, Chongqing, 400037, China. .,Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Army Military Medical University, Xinqiao Main Street, Chongqing, 400037, China. .,Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
80
|
Tyner JW, Haderk F, Kumaraswamy A, Baughn LB, Van Ness B, Liu S, Marathe H, Alumkal JJ, Bivona TG, Chan KS, Druker BJ, Hutson AD, Nelson PS, Sawyers CL, Willey CD. Understanding Drug Sensitivity and Tackling Resistance in Cancer. Cancer Res 2022; 82:1448-1460. [PMID: 35195258 PMCID: PMC9018544 DOI: 10.1158/0008-5472.can-21-3695] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/21/2022] [Accepted: 02/15/2022] [Indexed: 11/16/2022]
Abstract
Decades of research into the molecular mechanisms of cancer and the development of novel therapeutics have yielded a number of remarkable successes. However, our ability to broadly assign effective, rationally targeted therapies in a personalized manner remains elusive for many patients, and drug resistance persists as a major problem. This is in part due to the well-documented heterogeneity of cancer, including the diversity of tumor cell lineages and cell states, the spectrum of somatic mutations, the complexity of microenvironments, and immune-suppressive features and immune repertoires, which collectively require numerous different therapeutic approaches. Here, we describe a framework to understand the types and biological causes of resistance, providing translational opportunities to tackle drug resistance by rational therapeutic strategies.
Collapse
Affiliation(s)
- Jeffrey W. Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Franziska Haderk
- Department of Medicine, University of California, San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | | | - Linda B. Baughn
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Brian Van Ness
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Himangi Marathe
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Joshi J. Alumkal
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Trever G. Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - Keith Syson Chan
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, California
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Brian J. Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Alan D. Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Peter S. Nelson
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Charles L. Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Christopher D. Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
81
|
Tharmaseelan H, Hertel A, Tollens F, Rink J, Woźnicki P, Haselmann V, Ayx I, Nörenberg D, Schoenberg SO, Froelich MF. Identification of CT Imaging Phenotypes of Colorectal Liver Metastases from Radiomics Signatures-Towards Assessment of Interlesional Tumor Heterogeneity. Cancers (Basel) 2022; 14:cancers14071646. [PMID: 35406418 PMCID: PMC8997087 DOI: 10.3390/cancers14071646] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Tumoral heterogeneity (TH) is a major challenge in the treatment of metastatic colorectal cancer (mCRC) and is associated with inferior response. Therefore, the identification of TH would be beneficial for treatment planning. TH can be assessed by identifying genetic alterations. In this work, a radiomics-based approach for assessment of TH in colorectal liver metastases (CRLM) in CT scans is demonstrated. (2) Methods: In this retrospective study, CRLM of mCRC were segmented and radiomics features extracted using pyradiomics. Unsupervised k-means clustering was applied to features and lesions. Feature redundancy was evaluated by principal component analysis and reduced by Pearson correlation coefficient cutoff. Feature selection was conducted by LASSO regression and visual analysis of the clusters by radiologists. (3) Results: A total of 47 patients’ (36% female, median age 64) CTs with 261 lesions were included. Five clusters were identified, and the categories small disseminated (n = 31), heterogeneous (n = 105), homogeneous (n = 64), mixed (n = 59), and very large type (n = 2) were assigned based on visual characteristics. Further statistical analysis showed correlation (p < 0.01) of clusters with sex, primary location, T- and N-status, and mutational status. Feature reduction and selection resulted in the identification of four features as a final set for cluster definition. (4) Conclusions: Radiomics features can characterize TH in liver metastases of mCRC in CT scans, and may be suitable for a better pretherapeutic classification of liver lesion phenotypes.
Collapse
Affiliation(s)
- Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Alexander Hertel
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Fabian Tollens
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Johann Rink
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Piotr Woźnicki
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Verena Haselmann
- Institute of Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany;
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Dominik Nörenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Stefan O. Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Matthias F. Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
- Correspondence:
| |
Collapse
|
82
|
Kasi PM, Fehringer G, Taniguchi H, Starling N, Nakamura Y, Kotani D, Powles T, Li BT, Pusztai L, Aushev VN, Kalashnikova E, Sharma S, Malhotra M, Demko ZP, Aleshin A, Rodriguez A, Billings PR, Grothey A, Taieb J, Cunningham D, Yoshino T, Kopetz S. Impact of Circulating Tumor DNA-Based Detection of Molecular Residual Disease on the Conduct and Design of Clinical Trials for Solid Tumors. JCO Precis Oncol 2022; 6:e2100181. [PMID: 35263168 PMCID: PMC8926064 DOI: 10.1200/po.21.00181] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Earlier detection of cancer recurrence using circulating tumor DNA (ctDNA) to detect molecular residual disease (MRD) has the potential to dramatically affect cancer management. We review evidence supporting the use of ctDNA as a biomarker for detection of MRD and highlight the potential impact that ctDNA testing could have on the conduct of clinical trials.
Collapse
Affiliation(s)
- Pashtoon M Kasi
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | - Hiroya Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Naureen Starling
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Daisuke Kotani
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London ECMC, Barts Health, London, United Kingdom
| | - Bob T Li
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | | | | | | | | | | | | | | | | | - Axel Grothey
- West Cancer Center and Research Institute, Germantown, TN
| | - Julien Taieb
- Georges Pompidou European Hospital, SIRIC-CARPEM, Université de Paris, Paris, France
| | - David Cunningham
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
83
|
The potential of liquid biopsy in the management of cancer patients. Semin Cancer Biol 2022; 84:69-79. [PMID: 35331850 DOI: 10.1016/j.semcancer.2022.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
|
84
|
Strömberg C, Martinez de la Maza L, Fernández Moro C, Gerling M, Jorns C, Sparrelid E, Löhr J, Villard C. Prognostic impact of inter-metastatic heterogeneity of viable tumour cells in colorectal liver metastases. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 48:1656-1663. [DOI: 10.1016/j.ejso.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022]
|
85
|
Lee JK, Hazar-Rethinam M, Decker B, Gjoerup O, Madison RW, Lieber DS, Chung JH, Schrock AB, Creeden J, Venstrom J, Alexander B, Oxnard GR. The Pan-Tumor Landscape of Targetable Kinase Fusions in Circulating Tumor DNA. Clin Cancer Res 2022; 28:728-737. [PMID: 34753780 PMCID: PMC9377769 DOI: 10.1158/1078-0432.ccr-21-2136] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/15/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Oncogenic kinase fusions are targetable with approved and investigational therapies and can also mediate acquired resistance (AR) to targeted therapy. We aimed to understand the clinical validity of liquid biopsy comprehensive genomic profiling (CGP) to detect kinase fusions pan tumor. EXPERIMENTAL DESIGN CGP was performed on plasma and tissue samples during clinical care. All exons plus selected introns of 16 kinases involved in oncogenic fusions (ALK, BRAF, EGFR, ERBB2, FGFR1/2/3, MET, NTRK1/2/3, PDGFRA/B, RAF1, RET, and ROS1) were sequenced to capture fusions, including well-characterized and novel breakpoints. Plasma circulating tumor DNA (ctDNA) fraction was estimated to inform sensitivity. RESULTS Of 36,916 plasma cases, 32,492 (88%) had detectable ctDNA. Kinase fusions were detected in 1.8% of ctDNA-positive cases (571/32,492) and were most prevalent in patients with cholangiocarcinoma (4.2%), bladder cancer (3.6%), and non-small cell lung cancer (NSCLC; 3.1%). Of the 63 paired patient samples that had tissue and ctDNA specimens collected within 1 year and with estimated plasma ctDNA fraction >1%, fusions were detected in 47 of 51 (92%) liquid specimens with a fusion in the tissue sample. In 32 patients with fusions detected in liquid but not in tissue, 21 (66%) had evidence of putative acquired resistance. CONCLUSIONS Targetable kinase fusions are identified in ctDNA across cancer types. In pairs with tissue-identified fusions, fusion detection in ctDNA is reliable with elevated ctDNA fraction. These data support the validity of CGP to enable ctDNA-based fusion detection for informing clinical care in patients with advanced cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Geoffrey R. Oxnard
- Corresponding Author: Geoffrey R. Oxnard, Clinical Development, Foundation Medicine, Cambridge, MA 02141. Phone: 617-418-2200; E-mail:
| |
Collapse
|
86
|
Jacquemin V, Antoine M, Dom G, Detours V, Maenhaut C, Dumont JE. Dynamic Cancer Cell Heterogeneity: Diagnostic and Therapeutic Implications. Cancers (Basel) 2022; 14:280. [PMID: 35053446 PMCID: PMC8773841 DOI: 10.3390/cancers14020280] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Though heterogeneity of cancers is recognized and has been much discussed in recent years, the concept often remains overlooked in different routine examinations. Indeed, in clinical or biological articles, reviews, and textbooks, cancers and cancer cells are generally presented as evolving distinct entities rather than as an independent heterogeneous cooperative cell population with its self-oriented biology. There are, therefore, conceptual gaps which can mislead the interpretations/diagnostic and therapeutic approaches. In this short review, we wish to summarize and discuss various aspects of this dynamic evolving heterogeneity and its biological, pathological, clinical, diagnostic, and therapeutic implications, using thyroid carcinoma as an illustrative example.
Collapse
Affiliation(s)
- Valerie Jacquemin
- Correspondence: (V.J.); (J.E.D.); Tel.: +32-2-555-32-26 (V.J.); +32-2-555-41-34 (J.E.D.)
| | | | | | | | | | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.A.); (G.D.); (V.D.); (C.M.)
| |
Collapse
|
87
|
Giannini V, Pusceddu L, Defeudis A, Nicoletti G, Cappello G, Mazzetti S, Sartore-Bianchi A, Siena S, Vanzulli A, Rizzetto F, Fenocchio E, Lazzari L, Bardelli A, Marsoni S, Regge D. Delta-Radiomics Predicts Response to First-Line Oxaliplatin-Based Chemotherapy in Colorectal Cancer Patients with Liver Metastases. Cancers (Basel) 2022; 14:cancers14010241. [PMID: 35008405 PMCID: PMC8750408 DOI: 10.3390/cancers14010241] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Oxaliplatin-based chemotherapy remains the mainstay of first-line therapy in patients with metastatic colorectal cancer (mCRC). Unfortunately, only approximately 60% of treated patients achieve response, and half of responders will experience an early onset of disease progression. Furthermore, some individuals will develop a mixed response due to the emergence of resistant tumor subclones. The ability to predicting which patients will acquire resistance could help them avoid the unnecessary toxicity of oxaliplatin therapies. Furthermore, sorting out lesions that do not respond, in the context of an overall good response, could trigger further investigation into their mutational landscape, providing mechanistic insight towards the planning of a more comprehensive treatment. In this study, we validated a delta-radiomics signature capable of predicting response to oxaliplatin-based first-line treatment of individual liver colorectal cancer metastases. Findings could pave the way to a more personalized treatment of patients with mCRC. Abstract The purpose of this paper is to develop and validate a delta-radiomics score to predict the response of individual colorectal cancer liver metastases (lmCRC) to first-line FOLFOX chemotherapy. Three hundred one lmCRC were manually segmented on both CT performed at baseline and after the first cycle of first-line FOLFOX, and 107 radiomics features were computed by subtracting textural features of CT at baseline from those at timepoint 1 (TP1). LmCRC were classified as nonresponders (R−) if they showed progression of disease (PD), according to RECIST1.1, before 8 months, and as responders (R+), otherwise. After feature selection, we developed a decision tree statistical model trained using all lmCRC coming from one hospital. The final output was a delta-radiomics signature subsequently validated on an external dataset. Sensitivity, specificity, positive (PPV), and negative (NPV) predictive values in correctly classifying individual lesions were assessed on both datasets. Per-lesion sensitivity, specificity, PPV, and NPV were 99%, 94%, 95%, 99%, 85%, 92%, 90%, and 87%, respectively, in the training and validation datasets. The delta-radiomics signature was able to reliably predict R− lmCRC, which were wrongly classified by lesion RECIST as R+ at TP1, (93%, averaging training and validation set, versus 67% of RECIST). The delta-radiomics signature developed in this study can reliably predict the response of individual lmCRC to oxaliplatin-based chemotherapy. Lesions forecasted as poor or nonresponders by the signature could be further investigated, potentially paving the way to lesion-specific therapies.
Collapse
Affiliation(s)
- Valentina Giannini
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
- Correspondence:
| | - Laura Pusceddu
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Arianna Defeudis
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Giulia Nicoletti
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
| | - Giovanni Cappello
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Simone Mazzetti
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (A.S.-B.); (S.S.); (A.V.)
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (A.S.-B.); (S.S.); (A.V.)
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Angelo Vanzulli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (A.S.-B.); (S.S.); (A.V.)
- Department of Radiology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Francesco Rizzetto
- Department of Radiology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Luca Lazzari
- Precision Oncology, IFOM-The FIRC Institute of Molecular Oncology, 20139 Milan, Italy; (L.L.); (S.M.)
| | - Alberto Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, 10060 Candiolo, Italy;
- Department of Oncology, University of Torino, 10060 Candiolo, Italy
| | - Silvia Marsoni
- Precision Oncology, IFOM-The FIRC Institute of Molecular Oncology, 20139 Milan, Italy; (L.L.); (S.M.)
| | - Daniele Regge
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| |
Collapse
|
88
|
Kratz JD, Zhang W, Patel M, Uboha NV. Challenges in biomarker-based clinical trials for patients with gastrointestinal malignancies. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2022.2106852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Jeremy D. Kratz
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Division of Hematology, Medical Oncology and Palliative care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- US Department of Veterans Affairs, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Wei Zhang
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Monica Patel
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Division of Hematology, Medical Oncology and Palliative care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Nataliya V. Uboha
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Division of Hematology, Medical Oncology and Palliative care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
89
|
Mangiapane LR, Nicotra A, Turdo A, Gaggianesi M, Bianca P, Di Franco S, Sardina DS, Veschi V, Signore M, Beyes S, Fagnocchi L, Fiori ME, Bongiorno MR, Lo Iacono M, Pillitteri I, Ganduscio G, Gulotta G, Medema JP, Zippo A, Todaro M, De Maria R, Stassi G. PI3K-driven HER2 expression is a potential therapeutic target in colorectal cancer stem cells. Gut 2022; 71:119-128. [PMID: 33436496 PMCID: PMC8666826 DOI: 10.1136/gutjnl-2020-323553] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Cancer stem cells are responsible for tumour spreading and relapse. Human epidermal growth factor receptor 2 (HER2) expression is a negative prognostic factor in colorectal cancer (CRC) and a potential target in tumours carrying the gene amplification. Our aim was to define the expression of HER2 in colorectal cancer stem cells (CR-CSCs) and its possible role as therapeutic target in CRC resistant to anti- epidermal growth factor receptor (EGFR) therapy. DESIGN A collection of primary sphere cell cultures obtained from 60 CRC specimens was used to generate CR-CSC mouse avatars to preclinically validate therapeutic options. We also made use of the ChIP-seq analysis for transcriptional evaluation of HER2 activation and global RNA-seq to identify the mechanisms underlying therapy resistance. RESULTS Here we show that in CD44v6-positive CR-CSCs, high HER2 expression levels are associated with an activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, which promotes the acetylation at the regulatory elements of the Erbb2 gene. HER2 targeting in combination with phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase kinase (MEK) inhibitors induces CR-CSC death and regression of tumour xenografts, including those carrying Kras and Pik3ca mutation. Requirement for the triple targeting is due to the presence of cancer-associated fibroblasts, which release cytokines able to confer CR-CSC resistance to PI3K/AKT inhibitors. In contrast, targeting of PI3K/AKT as monotherapy is sufficient to kill liver-disseminating CR-CSCs in a model of adjuvant therapy. CONCLUSIONS While PI3K targeting kills liver-colonising CR-CSCs, the concomitant inhibition of PI3K, HER2 and MEK is required to induce regression of tumours resistant to anti-EGFR therapies. These data may provide a rationale for designing clinical trials in the adjuvant and metastatic setting.
Collapse
Affiliation(s)
- Laura Rosa Mangiapane
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Annalisa Nicotra
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Paola Bianca
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Davide Stefano Sardina
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | | | - Sven Beyes
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Luca Fagnocchi
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Micol Eleonora Fiori
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanita, Roma, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Melania Lo Iacono
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Irene Pillitteri
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Gloria Ganduscio
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Gaspare Gulotta
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, University of Amsterdam, Amsterdam, Noord-Holland, The Netherlands,Oncode Institute, University of Amsterdam, Amsterdam, Noord-Holland, The Netherlands
| | - Alessio Zippo
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Universita Cattolica del Sacro Cuore Facolta di Medicina e Chirurgia, Roma, Italy .,Policlinico A Gemelli, Roma, Lazio, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences, Università degli Studi di Palermo, Palermo, Italy
| |
Collapse
|
90
|
Ding S, Li H, Zhang YH, Zhou X, Feng K, Li Z, Chen L, Huang T, Cai YD. Identification of Pan-Cancer Biomarkers Based on the Gene Expression Profiles of Cancer Cell Lines. Front Cell Dev Biol 2021; 9:781285. [PMID: 34917619 PMCID: PMC8669964 DOI: 10.3389/fcell.2021.781285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
There are many types of cancers. Although they share some hallmarks, such as proliferation and metastasis, they are still very different from many perspectives. They grow on different organ or tissues. Does each cancer have a unique gene expression pattern that makes it different from other cancer types? After the Cancer Genome Atlas (TCGA) project, there are more and more pan-cancer studies. Researchers want to get robust gene expression signature from pan-cancer patients. But there is large variance in cancer patients due to heterogeneity. To get robust results, the sample size will be too large to recruit. In this study, we tried another approach to get robust pan-cancer biomarkers by using the cell line data to reduce the variance. We applied several advanced computational methods to analyze the Cancer Cell Line Encyclopedia (CCLE) gene expression profiles which included 988 cell lines from 20 cancer types. Two feature selection methods, including Boruta, and max-relevance and min-redundancy methods, were applied to the cell line gene expression data one by one, generating a feature list. Such list was fed into incremental feature selection method, incorporating one classification algorithm, to extract biomarkers, construct optimal classifiers and decision rules. The optimal classifiers provided good performance, which can be useful tools to identify cell lines from different cancer types, whereas the biomarkers (e.g. NCKAP1, TNFRSF12A, LAMB2, FKBP9, PFN2, TOM1L1) and rules identified in this work may provide a meaningful and precise reference for differentiating multiple types of cancer and contribute to the personalized treatment of tumors.
Collapse
Affiliation(s)
- ShiJian Ding
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Hao Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - XianChao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou, China
| | - ZhanDong Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Tao Huang
- CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
91
|
Rogers C, Morrissette JJD, Sussman RT. NTRK point mutations and their functional consequences. Cancer Genet 2021; 262-263:5-15. [PMID: 34972036 DOI: 10.1016/j.cancergen.2021.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/15/2021] [Accepted: 12/13/2021] [Indexed: 12/27/2022]
Abstract
The neurotrophic receptor tyrosine kinase (NTRK) family of genes, including NTRK1, NTRK2, and NTRK3, encodes membrane-bound receptors that normally regulate cell survival and differentiation upon binding of growth factors. Not surprisingly, mutations in these genes are known to contribute to the growth of a diverse number of cancers. With the recent FDA approval of two first-generation tyrosine-kinase inhibitors (TKIs) for adult and pediatric patients with solid tumors harboring NTRK gene fusions, much of the literature has focused on the biology behind these types of NTRK abnormalities; however, point mutations can also contribute to oncogenesis or resistance to TKI therapy, albeit at a lower frequency than fusions. This review focuses on NTRK gene mutations that are associated with resistance to directed therapies, mutations detected in the primary setting that confer increased oncogenic activity, and evidence that suggests that some of these variants may be treated using specific targeted therapies. Finally, this review focuses on the detection of point mutations, including the utility of cell-free DNA (cfDNA) for monitoring the acquisition of resistance mutations.
Collapse
Affiliation(s)
- Corey Rogers
- Department of Pathology and Laboratory Medicine, Division of Precision and Computational Diagnostics, University of Pennsylvania, 3020 Market Street, Suite 220, Philadelphia, PA 19104, USA.
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, Division of Precision and Computational Diagnostics, University of Pennsylvania, 3020 Market Street, Suite 220, Philadelphia, PA 19104, USA
| | - Robyn T Sussman
- Department of Pathology and Laboratory Medicine, Division of Precision and Computational Diagnostics, University of Pennsylvania, 3020 Market Street, Suite 220, Philadelphia, PA 19104, USA
| |
Collapse
|
92
|
Glennon KI, Maralani M, Abdian N, Paccard A, Montermini L, Nam AJ, Arseneault M, Staffa A, Jandaghi P, Meehan B, Brimo F, Tanguay S, Rak J, Riazalhosseini Y. Rational Development of Liquid Biopsy Analysis in Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13225825. [PMID: 34830979 PMCID: PMC8616270 DOI: 10.3390/cancers13225825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Among patients affected by renal cell carcinoma (RCC), the most common type of kidney cancer, it remains difficult to identify those who are at high risk for relapse or metastasis. This is in part due to the absence of reliable clinical biomarkers and robust methods to capture them. The aim of our study was to develop an improved assay to capture prognostic genomic biomarkers in circulating tumor DNA (ctDNA) in RCC. For this purpose, we first established a next generation sequencing (NGS) assay, targeting genes that are tailored for RCC and that are largely excluded from commercially available assays. Next, we showed the reliable performance of this assay to detect prognostic gene mutations in tumor DNA isolated from plasma, and from extracellular vesicles. Thus, our study provides a resource to facilitate ctDNA analysis for precision medicine in RCC. Abstract Renal cell carcinoma (RCC) is known for its variable clinical behavior and outcome, including heterogeneity in developing relapse or metastasis. Recent data highlighted the potential of somatic mutations as promising biomarkers for risk stratification in RCC. Likewise, the analysis of circulating tumor DNA (ctDNA) for such informative somatic mutations (liquid biopsy) is considered an important advance for precision oncology in RCC, allowing to monitor molecular disease evolution in real time. However, our knowledge about the utility of ctDNA analysis in RCC is limited, in part due to the lack of RCC-appropriate assays for ctDNA analysis. Here, by interrogating different blood compartments in xenograft models, we identified plasma cell-free (cf) DNA and extracellular vesicles (ev) DNA enriched for RCC-associated ctDNA. Additionally, we developed sensitive targeted sequencing and bioinformatics workflows capable of detecting somatic mutations in RCC-relevant genes with allele frequencies ≥ 0.5%. Applying this assay to patient-matched tumor and liquid biopsies, we captured tumor mutations in cf- and ev-DNA fractions isolated from the blood, highlighting the potentials of both fractions for ctDNA analysis. Overall, our study presents an RCC-appropriate sequencing assay and workflow for ctDNA analysis and provides a proof of principle as to the feasibility of detecting tumor-specific mutations in liquid biopsy in RCC patients.
Collapse
Affiliation(s)
- Kate I. Glennon
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
| | - Mahafarin Maralani
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Narges Abdian
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Antoine Paccard
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Laura Montermini
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Alice Jisoo Nam
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Madeleine Arseneault
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Alfredo Staffa
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Pouria Jandaghi
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
| | - Brian Meehan
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Fadi Brimo
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada;
| | - Simon Tanguay
- Division of Urology, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Janusz Rak
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Yasser Riazalhosseini
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
- Correspondence:
| |
Collapse
|
93
|
Yeoh Y, Low TY, Abu N, Lee PY. Regulation of signal transduction pathways in colorectal cancer: implications for therapeutic resistance. PeerJ 2021; 9:e12338. [PMID: 34733591 PMCID: PMC8544255 DOI: 10.7717/peerj.12338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Resistance to anti-cancer treatments is a critical and widespread health issue that has brought serious impacts on lives, the economy and public policies. Mounting research has suggested that a selected spectrum of patients with advanced colorectal cancer (CRC) tend to respond poorly to both chemotherapeutic and targeted therapeutic regimens. Drug resistance in tumours can occur in an intrinsic or acquired manner, rendering cancer cells insensitive to the treatment of anti-cancer therapies. Multiple factors have been associated with drug resistance. The most well-established factors are the emergence of cancer stem cell-like properties and overexpression of ABC transporters that mediate drug efflux. Besides, there is emerging evidence that signalling pathways that modulate cell survival and drug metabolism play major roles in the maintenance of multidrug resistance in CRC. This article reviews drug resistance in CRC as a result of alterations in the MAPK, PI3K/PKB, Wnt/β-catenin and Notch pathways.
Collapse
Affiliation(s)
- Yeelon Yeoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
94
|
Naidoo M, Piercey O, Tie J. Circulating Tumour DNA and Colorectal Cancer: the Next Revolutionary Biomarker? Curr Oncol Rep 2021; 23:140. [PMID: 34735665 DOI: 10.1007/s11912-021-01137-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Improving outcomes for patients with colorectal cancer in both the adjuvant and metastatic setting has been challenging. Here, we review the current and future directions for using ctDNA in clinical practice. RECENT FINDINGS Circulating tumour DNA (ctDNA) with its ability to detect minimal residual disease is beginning to refine the way we assess recurrence risk in the adjuvant setting. We can potentially tailor treatments to reduce recurrence risk and minimize treatment toxicity. In the metastatic setting, ctDNA can provide a less invasive method of detecting clinically important genetic changes to guide molecularly targeted treatment and to identify mechanisms of molecular resistance. ctDNA can be a surrogate marker for treatment response and help guide the timing of anti-EGFR rechallenge. We await the results of the randomized clinical trials assessing clinical utility of ctDNA in both the adjuvant and metastatic setting before incorporating ctDNA into clinical practice.
Collapse
Affiliation(s)
- Mahendra Naidoo
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Parkville Melbourne, VIC 3000, Australia.
| | - Oliver Piercey
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Parkville Melbourne, VIC 3000, Australia
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Parkville Melbourne, VIC 3000, Australia.,Division of Personalised Oncology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia.,Department of Medical Oncology, Western Health, Melbourne, VIC, 3021, Australia.,Sir Peter MacCallum, Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| |
Collapse
|
95
|
Ryu WK, Kim JS, Park MH, Lee M, Kim HJ, Ryu JS, Lim JH. Heterogeneous radiological response to chemotherapy is associated with poor prognosis in advanced non-small-cell lung cancer. Thorac Cancer 2021; 12:3333-3339. [PMID: 34693646 PMCID: PMC8671901 DOI: 10.1111/1759-7714.14207] [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: 08/25/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 11/30/2022] Open
Abstract
Background A heterogeneous radiological response is frequently observed in cancer patients and could reflect tumor heterogeneity. We investigated the prognostic impact of heterogeneous radiological responses in patients with advanced non‐small‐cell lung cancer (NSCLC) who received platinum‐based chemotherapy. Methods The treatment response according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria was evaluated in 212 patients with advanced NSCLC who received platinum‐based chemotherapy. Patients with partial response (PR) or stable disease (SD) were classified into “PR homo,” “PR hetero,” “SD homo,” and “SD hetero” by the presence of a heterogeneous radiological response, and survival was compared between groups. We also compared survival based on the presence of metabolic responses in lesions with heterogeneous radiological responses. Results Fifty‐two patients (24.5%) were classified as PR, 112 patients (52.8%) as SD, and 48 patients (22.7%) as progressive disease (PD). There was no significant difference in progression‐free survival (PFS) and overall survival (OS) between the PR homo and PR hetero groups. The SD homo group had a longer PFS and OS than the SD hetero group. In the SD hetero group, patients with increased maximum standardized uptake value (SUVmax) in lesions with heterogeneous radiological responses had a shorter PFS than those with a stable SUVmax. Conclusions The presence of lesions with radiological heterogeneity was associated with disease progression and poor prognosis in the SD group. Patients with heterogeneous radiological responses require careful monitoring.
Collapse
Affiliation(s)
- Woo Kyung Ryu
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jung Soo Kim
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Mi Hwa Park
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Minkyung Lee
- Department of Nuclear Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Hyun-Jung Kim
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jeong-Seon Ryu
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jun Hyeok Lim
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| |
Collapse
|
96
|
Bajgiran KR, Cordova AS, Elkhanoufi R, Dorman JA, Melvin AT. Simultaneous Droplet Generation with In-Series Droplet T-Junctions Induced by Gravity-Induced Flow. MICROMACHINES 2021; 12:mi12101211. [PMID: 34683262 PMCID: PMC8540845 DOI: 10.3390/mi12101211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 01/13/2023]
Abstract
Droplet microfluidics offers a wide range of applications, including high-throughput drug screening and single-cell DNA amplification. However, these platforms are often limited to single-input conditions that prevent them from analyzing multiple input parameters (e.g., combined cellular treatments) in a single experiment. Droplet multiplexing will result in higher overall throughput, lowering cost of fabrication, and cutting down the hands-on time in number of applications such as single-cell analysis. Additionally, while lab-on-a-chip fabrication costs have decreased in recent years, the syringe pumps required for generating droplets of uniform shape and size remain cost-prohibitive for researchers interested in utilizing droplet microfluidics. This work investigates the potential of simultaneously generating droplets from a series of three in-line T-junctions utilizing gravity-driven flow to produce consistent, well-defined droplets. Implementing reservoirs with equal heights produced inconsistent flow rates that increased as a function of the distance between the aqueous inlets and the oil inlet. Optimizing the three reservoir heights identified that taller reservoirs were needed for aqueous inlets closer to the oil inlet. Studying the relationship between the ratio of oil-to-water flow rates (Φ) found that increasing Φ resulted in smaller droplets and an enhanced droplet generation rate. An ANOVA was performed on droplet diameter to confirm no significant difference in droplet size from the three different aqueous inlets. The work described here offers an alternative approach to multiplexed droplet microfluidic devices allowing for the high-throughput interrogation of three sample conditions in a single device. It also has provided an alternative method to induce droplet formation that does not require multiple syringe pumps.
Collapse
|
97
|
Inferring Copy Number from Triple-Negative Breast Cancer Patient Derived Xenograft scRNAseq Data Using scCNA. Methods Mol Biol 2021. [PMID: 34590283 DOI: 10.1007/978-1-0716-1740-3_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Cancer can develop from an accumulation of alterations, some of which cause a nonmalignant cell to transform to a malignant state exhibiting increased rate of cell growth and evasion of growth suppressive mechanisms, eventually leading to tissue invasion and metastatic disease. Triple-negative breast cancers (TNBC) are heterogeneous and are clinically characterized by the lack of expression of hormone receptors and human epidermal growth factor receptor 2 (HER2), which limits its treatment options. Since tumor evolution is driven by diverse cancer cell populations and their microenvironment, it is imperative to map TNBC at single-cell resolution. Here, we describe an experimental procedure for isolating a single-cell suspension from a TNBC patient-derived xenograft, subjecting it to single-cell RNA sequencing using droplet-based technology from 10× Genomics and analyzing the transcriptomic data at single-cell resolution to obtain inferred copy number aberration profiles, using scCNA. Data obtained using this single-cell RNA sequencing experimental and analytical methodology should enhance our understanding of intratumor heterogeneity which is key for identifying genetic vulnerabilities and developing effective therapies.
Collapse
|
98
|
Increased sensitivity to SMAC mimetic LCL161 identified by longitudinal ex vivo pharmacogenomics of recurrent, KRAS mutated rectal cancer liver metastases. J Transl Med 2021; 19:384. [PMID: 34496878 PMCID: PMC8424985 DOI: 10.1186/s12967-021-03062-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022] Open
Abstract
Tumor heterogeneity is a primary cause of treatment failure. However, changes in drug sensitivity over time are not well mapped in cancer. Patient-derived organoids (PDOs) may predict clinical drug responses ex vivo and offer an opportunity to evaluate novel treatment strategies in a personalized fashion. Here we have evaluated spatio-temporal functional and molecular dynamics of five PDO models established after hepatic re-resections and neoadjuvant combination chemotherapies in a patient with microsatellite stable and KRAS mutated metastatic rectal cancer. Histopathological differentiation phenotypes of the PDOs corresponded with the liver metastases, and ex vivo drug sensitivities generally reflected clinical responses and selection pressure, assessed in comparison to a reference data set of PDOs from metastatic colorectal cancers. PDOs from the initial versus the two recurrent metastatic settings showed heterogeneous cell morphologies, protein marker expression, and drug sensitivities. Exploratory analyses of a drug screen library of 33 investigational anticancer agents showed the strongest ex vivo sensitivity to the SMAC mimetic LCL161 in PDOs of recurrent disease compared to those of the initial metastasis. Functional analyses confirmed target inhibition and apoptosis induction in the LCL161 sensitive PDOs from the recurrent metastases. Gene expression analyses indicated an association between LCL161 sensitivity and tumor necrosis factor alpha signaling and RIPK1 gene expression. In conclusion, LCL161 was identified as a possible experimental therapy of a metastatic rectal cancer that relapsed after hepatic resection and standard systemic treatment.
Collapse
|
99
|
Drenner K, Basu GD, Goodman LJ, Ozols AA, LoBello JR, Royce T, Gordon MS, Borazanci EH, Steinbach MA, Trent J, Sharma S. The value of comprehensive genomic sequencing to maximize the identification of clinically actionable alterations in advanced cancer patients: a case series. Oncotarget 2021; 12:1836-1847. [PMID: 34504655 PMCID: PMC8416559 DOI: 10.18632/oncotarget.28046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023] Open
Abstract
PURPOSE We present seven cases of advanced cancer patients who initially underwent tumor testing utilizing smaller, panel-based tests, followed by a variety of therapeutic treatments which ultimately resulted in progression of their disease. These cases demonstrate the value of utilizing WES/RNA seq and characterization following disease progression in these patients and the determination of clinically targetable alterations as well as acquired resistance mutations. MATERIALS AND METHODS All patients are part of an IRB approved observational study. WES and RNA sequencing were performed, using GEM ExTra® on tumor and blood samples obtained during routine clinical care. To accurately determine somatic versus germline alterations the test was performed with paired normal testing from peripheral blood. RESULTS The presented cases demonstrate the clinical impact of actionable findings uncovered using GEM ExTra® in patients with advanced disease who failed many rounds of treatment. Unique alterations were identified resulting in newly identified potential targeted therapies, mechanisms of resistance, and variation in the genomic characterization of the primary versus the metastatic tumor. CONCLUSIONS Taken together our results demonstrate that GEM ExTra® maximizes detection of actionable mutations, thus allowing for appropriate treatment selection for patients harboring both common and rare genomic alterations.
Collapse
Affiliation(s)
- Kevin Drenner
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
- These authors contributed equally to this work
| | - Gargi D. Basu
- Ashion Analytics, LLC, Phoenix, AZ 85004, USA
- These authors contributed equally to this work
| | | | | | | | | | | | | | | | - Jeffrey Trent
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
| | - Sunil Sharma
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
| |
Collapse
|
100
|
Zhuang QS, Sun XB, Chong QY, Banerjee A, Zhang M, Wu ZS, Zhu T, Pandey V, Lobie PE. ARTEMIN Promotes Oncogenicity and Resistance to 5-Fluorouracil in Colorectal Carcinoma by p44/42 MAPK Dependent Expression of CDH2. Front Oncol 2021; 11:712348. [PMID: 34422665 PMCID: PMC8377398 DOI: 10.3389/fonc.2021.712348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
ARTEMIN (ARTN), one of the glial-cell derived neurotrophic factor family of ligands, has been reported to be associated with a number of human malignancies. In this study, the enhanced expression of ARTN in colorectal carcinoma (CRC) was observed; the expression of ARTN positively correlated with lymph node metastases and advanced tumor stages and predicted poor prognosis. Forced expression of ARTN in CRC cells enhanced oncogenic behavior, mesenchymal phenotype, stem cell-like properties and tumor growth and metastasis in a xenograft model. These functions were conversely inhibited by depletion of endogenous ARTN. Forced expression of ARTN reduced the sensitivity of CRC cells to 5-FU treatment; and 5-FU resistant CRC cells harbored enhanced expression of ARTN. The oncogenic functions of ARTN were demonstrated to be mediated by p44/42 MAP kinase dependent expression of CDH2 (CADHERIN 2, also known as N-CADHERIN). Inhibition of p44/42 MAP kinase activity or siRNA mediated depletion of endogenous CDH2 reduced the enhanced oncogenicity and chemoresistance consequent to forced expression of ARTN induced cell functions; and forced expression of CDH2 rescued the reduced mesenchymal properties and resistance to 5-FU after ARTN depletion. In conclusion, ARTN may be of prognostic and theranostic utility in CRC.
Collapse
Affiliation(s)
- Qiu-Shi Zhuang
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xin-Bao Sun
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore
| | - Arindam Banerjee
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Department of Chemical Engineering, Indian Institute of Technology, Kharagpur, India
| | - Min Zhang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Tao Zhu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|