1
|
Sheffield BS, Banerji S, Chankowsky A, Dudani S, Gill S, Gorski Z, Kassam S, Macaulay C, Manna M, Perdrizet K, Ramjeesingh R, D’Angelo MS, Servidio-Italiano F. Toward Timely and Equitable Advanced Biomarker Testing for Patients with Metastatic Cancer in Canada. Curr Oncol 2025; 32:141. [PMID: 40136345 PMCID: PMC11941070 DOI: 10.3390/curroncol32030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
The explosion in biomarker testing over the past two decades continues to transform cancer care in Canada and around the world. Precision medicine is supported by identifying actionable mutations that direct therapeutic choices, thus improving survival and quality of life, especially for patients with advanced/metastatic disease. In addition, our growing understanding of the genetic basis of cancer is advanced by research employing ever-expanding databases of genetic mutations, therapies and outcomes. Despite this promising progress, however, access to biomarker testing remains inequitable across Canada, to the detriment of patients. Several underlying factors contribute to this situation, including the need for investment in and standardization of laboratory medicine infrastructure and processes, and the lack of suitable methods for cost/benefit evaluations to inform funding decisions. In 2024, a Canadian conference brought together patients, clinicians, researchers, policy-makers and scientists to address "Equitable Access to Advanced Biomarker Testing for Canadian Metastatic Cancer Patients". Two major themes arose from the conference: the urgent need to adopt comprehensive genomic profiling (CGP) as a standard of care across Canada, and the emerging role of liquid biopsy in accelerating access to biomarker testing for patients with advanced/metastatic cancer.
Collapse
Affiliation(s)
- Brandon S. Sheffield
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (B.S.S.); (S.D.); (K.P.)
| | - Shantanu Banerji
- Rady Faculty of Health Sciences, Department of Medical Oncology and Hematology, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Allen Chankowsky
- Patient Advocate, Member of the ASCO TAPUR molecular tumour board, Toronto, ON, Canada;
| | - Shaan Dudani
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (B.S.S.); (S.D.); (K.P.)
| | | | - Zuzanna Gorski
- Anatomical Pathology and Laboratory Medicine, QEII Health Sciences Centre, Halifax, NS B3H 1V8, Canada;
| | - Shaqil Kassam
- Stronach Regional Cancer Centre, Newmarket, ON L3Y 2P9, Canada;
| | - Cassandra Macaulay
- Colorectal Cancer Resource & Action Network (CCRAN), Toronto, ON M4W 3E2, Canada; (C.M.); (M.S.D.)
| | - Mita Manna
- Saskatoon Cancer Centre, Saskatoon, SK S7N 4H4, Canada;
| | - Kirstin Perdrizet
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (B.S.S.); (S.D.); (K.P.)
| | - Ravi Ramjeesingh
- Division of Medical Oncology, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada;
| | - Monika Slovinec D’Angelo
- Colorectal Cancer Resource & Action Network (CCRAN), Toronto, ON M4W 3E2, Canada; (C.M.); (M.S.D.)
| | | |
Collapse
|
2
|
Maher MH, Duose DY, Wistuba II, Luthra R, Arjuna S, Roy-Chowdhuri S. A rapid turnaround time workflow for a cytological liquid biopsy assay using FNA supernatant specimens. Cancer Cytopathol 2025; 133:e22925. [PMID: 39704279 DOI: 10.1002/cncy.22925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/03/2024] [Accepted: 10/23/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Genomic profiling is essential in the management of non-small cell lung cancer. However, this may often be challenging because of limited cytological tissue and extended turnaround time (TAT) for next-generation sequencing (NGS). This study aims to describe a rapid TAT workflow for molecular profiling using fine-needle aspiration (FNA) supernatants. METHODS A fully automated total nucleic acid extraction using the Genexus Integrated System was compared to a manual extraction using FNA supernatants from 50 patients with non-small cell lung cancer. Molecular profiling using the 50 gene Oncomine Precision Assay GX panel was performed and NGS results were compared with those of paired tissue samples. RESULTS The FNA samples processed using the automated Genexus purification system (n = 42) and the manual extraction method (n = 8) showed comparable quality control metrics with a median total nucleic acid yield of 1230 ng (18-17720 ng) and 1068 ng (777-1740 ng), respectively. The Genexus purification system reduced the hands-on time from 120 to 30 minutes, enhancing workflow efficiency and decreasing the overall TAT. Of the 50 samples extracted, NGS was performed on 26 samples: seven via manual extraction and 19 using automated extraction. NGS quality control metrics were also comparable between both extraction methods. The overall TAT of the automated NGS workflow from specimen received to test result was 24 hours, providing rapid and reliable molecular results for timely clinical decision-making and improved patient outcomes.
Collapse
Affiliation(s)
- Mohamed H Maher
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- South Egypt Cancer Institute-Assiut University, Houston, Texas, USA
| | - Dzifa Y Duose
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ignacio I Wistuba
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Srividya Arjuna
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
3
|
Bestvina CM, Waters D, Morrison L, Emond B, Lafeuille MH, Hilts A, Mujwara D, Lefebvre P, He A, Vanderpoel J. Impact of next-generation sequencing vs polymerase chain reaction testing on payer costs and clinical outcomes throughout the treatment journeys of patients with metastatic non-small cell lung cancer. J Manag Care Spec Pharm 2024; 30:1467-1478. [PMID: 39259000 DOI: 10.18553/jmcp.2024.24137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
BACKGROUND For patients with metastatic non-small cell lung cancer (mNSCLC), next-generation sequencing (NGS) biomarker testing has been associated with a faster time to appropriate targeted therapy and more comprehensive testing relative to polymerase chain reaction (PCR) testing. However, the impact on payer costs and clinical outcomes during patients' treatment journeys has not been fully characterized. OBJECTIVE To assess the costs and clinical outcomes of NGS vs PCR biomarker testing among patients with newly diagnosed de novo mNSCLC from a US payers' perspective. METHODS A Markov model assessed costs and clinical outcomes of NGS vs PCR testing from the start of testing up to 3 years after. Patients entered the model after receiving biomarker test results and then initiated first-line (1L) targeted or nontargeted therapy (immunotherapy and/or chemotherapy) depending on actionable mutation detection. A few patients with an actionable mutation were not detected by PCR and inappropriately initiated 1L nontargeted therapy. At each 1-month cycle, patients could remain on treatment with 1L, progress to second line or later (2L+), or die. Literature-based inputs included the rates of progression-free survival (PFS) and overall survival (OS), targeted and nontargeted therapy costs, total costs of testing, and medical costs of 1L, 2L+, and death. Per patient average PFS and OS as well as cumulative costs were reported for NGS and PCR testing. RESULTS In a modeled population of 100 patients (75% commercial and 25% Medicare), 45.9% of NGS and 40.0% of PCR patients tested positive for an actionable mutation. Relative to PCR, NGS was associated with $7,386 in savings per patient (NGS = $326,154; PCR = $333,540) at 1 year, driven by lower costs of testing, including estimated costs of delayed care and nontargeted therapy initiation before receiving test results (NGS = $8,866; PCR = $16,373). Treatment costs were similar (NGS = $305,644; PCR = $305,283). In the PCR cohort, the per patient costs of inappropriate 1L nontargeted therapy owing to undetected mutations were $6,455, $6,566, and $6,569 over the first 1, 2, and 3 years, respectively. Relative to PCR testing, NGS was associated with $4,060 in savings at 2 years and $1,092 at 3 years. Patients who initiated 1L targeted therapy had an additional 5.4, 8.8, and 10.4 months of PFS and an additional 1.4, 3.6, and 5.3 months of OS over the first 1, 2, and 3 years, respectively, relative to those who inappropriately initiated 1L nontargeted therapy. CONCLUSIONS In this Markov model, as early as year 1, and over 3 years following biomarker testing, patients with newly diagnosed de novo mNSCLC undergoing NGS testing are projected to have cost savings and longer PFS and OS relative to those tested with PCR.
Collapse
Affiliation(s)
| | - Dexter Waters
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| | | | | | | | | | | | | | - Andy He
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| | - Julie Vanderpoel
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| |
Collapse
|
4
|
Breadner D, Hwang DM, Husereau D, Cheema P, Doucette S, Ellis PM, Kassam S, Leighl N, Maziak DE, Selvarajah S, Sheffield BS, Juergens RA. Implementation of Liquid Biopsy in Non-Small-Cell Lung Cancer: An Ontario Perspective. Curr Oncol 2024; 31:6017-6031. [PMID: 39451753 PMCID: PMC11505603 DOI: 10.3390/curroncol31100449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in Canada, with non-small-cell lung cancer (NSCLC) accounting for the majority of cases. Timely access to comprehensive molecular profiling is critical for selecting biomarker-matched targeted therapies, which lead to improved outcomes in advanced NSCLC. Tissue biopsy samples are the gold standard for molecular profiling; however, several challenges can prevent timely and complete molecular profiling from being performed, causing delays in treatment or suboptimal therapy selection. Liquid biopsy offers a minimally invasive method for molecular profiling by analyzing circulating tumour DNA (ctDNA) and RNA (cfRNA) in plasma, potentially overcoming these barriers. This paper discusses the outcomes of a multidisciplinary working group in Ontario, which proposed three eligibility criteria for liquid biopsy reimbursement: (1) insufficient tissue for complete testing or failed tissue biomarker testing; (2) suspected advanced NSCLC where tissue biopsy is not feasible; and (3) high-risk patients who may deteriorate before tissue results are available. The group also addressed considerations for assay selection, implementation, and economic impact. These discussions aim to inform reimbursement and implementation strategies for liquid biopsy in Ontario's public healthcare system, recognizing the need for ongoing evaluation as technology and evidence evolve.
Collapse
Affiliation(s)
- Daniel Breadner
- Verspeeten Family Cancer Centre, London Health Sciences Center, London, ON N6A 5W9, Canada
| | - David M. Hwang
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Don Husereau
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Parneet Cheema
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Peter M. Ellis
- Division of Medical Oncology, Juravinski Cancer Centre, Hamilton, ON L8V 5C2, Canada (R.A.J.)
- Department of Oncology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shaqil Kassam
- Southlake Stronach Regional Cancer Centre, Newmarket, ON L3Y 2P9, Canada
| | - Natasha Leighl
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Donna E. Maziak
- Department of Thoracic Surgery, The Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
| | - Shamini Selvarajah
- Laboratory Medicine Program, Division of Genome Diagnostics, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Brandon S. Sheffield
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Rosalyn A. Juergens
- Division of Medical Oncology, Juravinski Cancer Centre, Hamilton, ON L8V 5C2, Canada (R.A.J.)
- Department of Oncology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
5
|
Stewart DJ. Has the blood-brain barrier finally been busted? Oncologist 2024; 29:645-647. [PMID: 38837042 PMCID: PMC11299943 DOI: 10.1093/oncolo/oyae131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Faith in the blood-brain barrier has been remarkably resilient. This commentary questions its importance in the treatment of brain metastases.
Collapse
|
6
|
Nicholas C, Beharry A, Bendzsak AM, Bisson KR, Dadson K, Dudani S, Iafolla M, Irshad K, Perdrizet K, Raskin W, Singh R, Tsui DCC, Wang X, Yeung C, Cheema PK, Sheffield BS. Point of Care Liquid Biopsy for Cancer Treatment-Early Experience from a Community Center. Cancers (Basel) 2024; 16:2505. [PMID: 39061145 PMCID: PMC11274424 DOI: 10.3390/cancers16142505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Liquid biopsy is rapidly becoming an indispensable tool in the oncologist's arsenal; however, this technique remains elusive in a publicly funded healthcare system, and real-world evidence is needed to demonstrate utility and feasibility. Here, we describe the first experience of an in-house point of care liquid biopsy program at a Canadian community hospital. A retrospective review of consecutive cases that underwent plasma-based next-generation sequencing (NGS) was conducted. Liquid biopsy was initiated at the discretion of clinicians. Sequencing followed a point of care workflow using the Genexus™ integrated sequencer and the Oncomine precision assay, performed by histotechnologists. Results were reported by the attending pathologist. Eligible charts were reviewed for outcomes of interest, including the intent of the liquid biopsy, results of the liquid biopsy, and turnaround time from blood draw to results available. A total of 124 cases, with confirmed or suspected cancer, underwent liquid biopsy between January 2021 and November 2023. The median turnaround time for liquid biopsy results was 3 business days (range 1-12 days). The sensitivity of liquid biopsies was 71%, compared to tissue testing in cases with matched tissue results available for comparison. Common mutations included EGFR (29%), in 86 lung cancer patients, and PIK3CA (22%), identified in 13 breast cancer patients. Healthcare providers ordered liquid biopsies to inform diagnostic investigations and treatment decisions, and to determine progression or resistance mechanisms, as these reasons often overlapped. This study demonstrates that rapid in-house liquid biopsy using point of care methodology is feasible. The technique facilitates precision treatment and offers many additional advantages for cancer care.
Collapse
Affiliation(s)
- Champica Nicholas
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Andrea Beharry
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Anna M. Bendzsak
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Kassandra R. Bisson
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Keith Dadson
- Thermo Fisher Scientific, Burlington, ON L7L 5Z1, Canada
| | - Shaan Dudani
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Marco Iafolla
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Kashif Irshad
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Kirstin Perdrizet
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - William Raskin
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Raviya Singh
- Division of Medical Oncology, Scarborough Health Network, Scarborough, ON M1P 2V5, Canada
| | - David Chun Cheong Tsui
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Xin Wang
- Division of Medical Oncology, UHN Princess Margaret Cancer Centre, Toronto, ON M5S 1A1, Canada
| | - Ching Yeung
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Parneet K. Cheema
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Brandon S. Sheffield
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| |
Collapse
|
7
|
Stewart DJ, Cole K, Bosse D, Brule S, Fergusson D, Ramsay T. Population Survival Kinetics Derived from Clinical Trials of Potentially Curable Lung Cancers. Curr Oncol 2024; 31:1600-1617. [PMID: 38534955 PMCID: PMC10968953 DOI: 10.3390/curroncol31030122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 05/26/2024] Open
Abstract
Using digitized data from progression-free survival (PFS) and overall survival Kaplan-Meier curves, one can assess population survival kinetics through exponential decay nonlinear regression analyses. To demonstrate their utility, we analyzed PFS curves from published curative-intent trials of non-small cell lung cancer (NSCLC) adjuvant chemotherapy, adjuvant osimertinib in resected EGFR-mutant NSCLC (ADAURA trial), chemoradiotherapy for inoperable NSCLC, and limited small cell lung cancer (SCLC). These analyses permit assessment of log-linear curve shape and estimation of the proportion of patients cured, PFS half-lives for subpopulations destined to eventually relapse, and probability of eventual relapse in patients remaining progression-free at different time points. The proportion of patients potentially cured was 41% for adjuvant controls, 58% with adjuvant chemotherapy, 17% for ADAURA controls, not assessable with adjuvant osimertinib, 15% with chemoradiotherapy, and 12% for SCLC. Median PFS half-life for relapsing subpopulations was 11.9 months for adjuvant controls, 17.4 months with adjuvant chemotherapy, 24.4 months for ADAURA controls, not assessable with osimertinib, 9.3 months with chemoradiotherapy, and 10.7 months for SCLC. For those remaining relapse-free at 2 and 5 years, the cure probability was 74%/96% for adjuvant controls, 77%/93% with adjuvant chemotherapy, 51%/94% with chemoradiation, and 39%/87% with limited SCLC. Relatively easy population kinetic analyses add useful information.
Collapse
Affiliation(s)
- David J. Stewart
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Katherine Cole
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | - Dominick Bosse
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Stephanie Brule
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Dean Fergusson
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Tim Ramsay
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| |
Collapse
|
8
|
Grafham GK, Craddock KJ, Huang W, Louie AV, Zhang L, Hwang DM, Parmar A. Referred molecular testing as a barrier to optimal treatment decision making in metastatic non-small cell lung cancer: Experience at a tertiary academic institution in Canada. Cancer Med 2024; 13:e6886. [PMID: 38317584 PMCID: PMC10905241 DOI: 10.1002/cam4.6886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/08/2023] [Accepted: 12/16/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Molecular testing is critical to guiding treatment approaches in patients with metastatic non-small cell lung cancer (mNSCLC), with testing delays adversely impacting the timeliness of treatment decisions. Here, we aimed to evaluate the time from initial mNSCLC diagnosis to treatment decision (TTD) following implementation of in-house EGFR, ALK, and PD-L1 testing at our institution. METHODS We conducted a retrospective chart review of 165 patients (send-out testing, n = 92; in-house testing, n = 73) with newly diagnosed mNSCLC treated at our institution. Data were compared during the send-out (March 2017-May 2019) and in-house (July 2019-March 2021) testing periods. We performed a detailed workflow analysis to provide insight on the pre-analytic, analytic, and post-analytic intervals that constituted the total TTD. RESULTS TTD was significantly shorter with in-house testing (10 days vs. 18 days, p < 0.0001), driven largely by decreased internal handling and specimen transit times (2 days vs. 3 days, p < 0.0001) and laboratory turnaround times (TAT, 3 days vs. 8 days, p < 0.0001), with 96% of in-house cases meeting the international guideline of a ≤ 10-day intra-laboratory TAT (vs. 74% send-out, p < 0.001). Eighty-eight percent of patients with in-house testing had results available at their first oncology consultation (vs. 52% send-out, p < 0.0001), and all patients with in-house testing had results available at the time of treatment decision (vs. 86% send-out, p = 0.57). CONCLUSION Our results demonstrate the advantages of in-house biomarker testing for mNSCLC at a tertiary oncology center. Incorporation of in-house testing may reduce barriers to offering personalized medicine by improving the time to optimal systemic therapy decision.
Collapse
Affiliation(s)
- Grace K. Grafham
- Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Kenneth J. Craddock
- Department of Laboratory Medicine and Molecular DiagnosticsSunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Weei‐Yuarn Huang
- Department of Laboratory Medicine and Molecular DiagnosticsSunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Alexander V. Louie
- Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
- Department of Radiation OncologySunnybrook Health Sciences CentreTorontoOntarioCanada
| | | | - David M. Hwang
- Department of Laboratory Medicine and Molecular DiagnosticsSunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Ambica Parmar
- Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
- Division of Hematology and Medical Oncology, Department of MedicineSunnybrook Health Sciences CentreTorontoOntarioCanada
| |
Collapse
|
9
|
Bestvina CM, Waters D, Morrison L, Emond B, Lafeuille MH, Hilts A, Lefebvre P, He A, Vanderpoel J. Cost of genetic testing, delayed care, and suboptimal treatment associated with polymerase chain reaction versus next-generation sequencing biomarker testing for genomic alterations in metastatic non-small cell lung cancer. J Med Econ 2024; 27:292-303. [PMID: 38391239 DOI: 10.1080/13696998.2024.2314430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
AIMS To assess US payers' per-patient cost of testing associated with next-generation sequencing (NGS) versus polymerase chain reaction (PCR) biomarker testing strategies among patients with metastatic non-small cell lung cancer (mNSCLC), including costs of testing, delayed care, and suboptimal treatment initiation. METHODS A decision tree model considered biomarker testing for genomic alterations using either NGS, sequential PCR testing, or hotspot panel PCR testing. Literature-based model inputs included time-to-test results, costs for testing/medical care, costs of delaying care, costs of immunotherapy [IO]/chemotherapy [CTX] initiation prior to receiving test results, and costs of suboptimal treatment initiation after test results (i.e. costs of first-line IO/CTX in patients with actionable mutations that were undetected by PCR that would have been identified with NGS). The proportion of patients testing positive for a targetable alteration, time to appropriate therapy initiation, and per-patient costs were estimated for NGS and PCR strategies combined. RESULTS In a modeled cohort of 1,000,000 members (25% Medicare, 75% commercial), an estimated 1,119 had mNSCLC and received testing. The proportion of patients testing positive for a targetable alteration was 45.9% for NGS and 40.0% for PCR testing. Mean per-patient costs were lowest for NGS ($8,866) compared to PCR ($18,246), with lower delayed care costs of $1,301 for NGS compared to $3,228 for PCR, and lower costs of IO/CTX initiation prior to receiving test results (NGS: $2,298; PCR:$5,991). Cost savings, reaching $10,496,220 at the 1,000,000-member plan level, were driven by more rapid treatment with appropriate therapy for patients tested with NGS (2.1 weeks) compared to PCR strategies (5.2 weeks). LIMITATIONS Model inputs/assumptions were based on published literature or expert opinion. CONCLUSIONS NGS testing was associated with greater cost savings versus PCR, driven by more rapid results, shorter time to appropriate therapy initiation, and minimized use of inappropriate therapies while awaiting and after test results.
Collapse
Affiliation(s)
- Christine M Bestvina
- University of Chicago Comprehensive Cancer Center; Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Dexter Waters
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
| | | | | | | | | | | | - Andy He
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
| | - Julie Vanderpoel
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
| |
Collapse
|
10
|
Silas U, Blüher M, Bosworth Smith A, Saunders R. Fast In-House Next-Generation Sequencing in the Diagnosis of Metastatic Non-small Cell Lung Cancer: A Hospital Budget Impact Analysis. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2023; 10:111-118. [PMID: 37389301 PMCID: PMC10306161 DOI: 10.36469/001c.77686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Background: Targeted therapy for cancer is becoming more frequent as the understanding of the molecular pathogenesis increases. Molecular testing must be done to use targeted therapy. Unfortunately, the testing turnaround time can delay the initiation of targeted therapy. Objective: To investigate the impact of a next-generation sequencing (NGS) machine in the hospital that would allow for in-house NGS testing of metastatic non-small cell lung cancer (mNSCLC) in a US setting. Methods: The differences between 2 hospital pathways were established with a cohort-level decision tree that feeds into a Markov model. A pathway that used in-house NGS (75%) and the use of external laboratories (so-called send-out NGS) (25%), was compared with the standard of exclusively send-out NGS. The model was from the perspective of a US hospital over a 5-year time horizon. All cost input data were in or inflated to 2021 USD. Scenario analysis was done on key variables. Results: In a hospital with 500 mNSCLC patients, the implementation of in-house NGS was estimated to increase the testing costs and the revenue of the hospital. The model predicted a $710 060 increase in testing costs, a $1 732 506 increase in revenue, and a $1 022 446 return on investment over 5 years. The payback period was 15 months with in-house NGS. The number of patients on targeted therapy increased by 3.38%, and the average turnaround time decreased by 10 days when in-house NGS was used. Discussion: Reducing testing turnaround time is a benefit of in-house NGS. It could contribute to fewer mNSCLC patients lost to second opinion and an increased number of patients on targeted therapy. The model outcomes predicted that, over a 5-year period, there would be a positive return on investment for a US hospital. The model reflects a proposed scenario. The heterogeneity of hospital inputs and the cost of send-out NGS means context-specific inputs are needed. Conclusion: Using in-house NGS testing could reduce the testing turnaround time and increase the number of patients on targeted therapy. Additional benefits for the hospital are that fewer patients will be lost to second opinion and that in-house NGS could generate additional revenue.
Collapse
Affiliation(s)
- Ubong Silas
- Coreva Scientific GmbH & Co. KG, Königswinter, Germany
| | | | | | | |
Collapse
|
11
|
Sheffield BS, Eaton K, Emond B, Lafeuille MH, Hilts A, Lefebvre P, Morrison L, Stevens AL, Ewara EM, Cheema P. Cost Savings of Expedited Care with Upfront Next-Generation Sequencing Testing versus Single-Gene Testing among Patients with Metastatic Non-Small Cell Lung Cancer Based on Current Canadian Practices. Curr Oncol 2023; 30:2348-2365. [PMID: 36826141 PMCID: PMC9955559 DOI: 10.3390/curroncol30020180] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
This study assessed the total costs of testing, including the estimated costs of delaying care, associated with next-generation sequencing (NGS) versus single-gene testing strategies among patients with newly diagnosed metastatic non-small cell lung cancer (mNSCLC) from a Canadian public payer perspective. A decision tree model considered testing for genomic alterations using tissue biopsy NGS or single-gene strategies following Canadian guideline recommendations. Inputs included prevalence of mNSCLC, the proportion that tested positive for each genomic alteration, rebiopsy rates, time to test results, testing/medical costs, and costs of delaying care based on literature, public data, and expert opinion. Among 1,000,000 hypothetical publicly insured adult Canadians (382 with mNSCLC), the proportion of patients that tested positive for a genomic alteration with an approved targeted therapy was 38.0% for NGS and 26.1% for single-gene strategies. The estimated mean time to appropriate targeted therapy initiation was 5.1 weeks for NGS and 9.2 weeks for single-gene strategies. Based on literature, each week of delayed care cost CAD 406, translating to total mean per-patient costs of CAD 3480 for NGS and CAD 5632 for single-gene strategies. NGS testing with mNSCLC in current Canadian practice resulted in more patients with an identified mutation, shorter time to appropriate targeted therapy initiation, and lower total testing costs compared to single-gene strategies.
Collapse
Affiliation(s)
| | | | - Bruno Emond
- Analysis Group, Inc., Montréal, QC H3B 0G7, Canada
| | | | | | | | - Laura Morrison
- Analysis Group, Inc., Montréal, QC H3B 0G7, Canada
- Correspondence: ; Tel.: +514-871-3303
| | | | | | - Parneet Cheema
- William Osler Health System, Brampton, ON L6R 3J7, Canada
| |
Collapse
|
12
|
Fung-Kee-Fung M, Ozer RS, Davies B, Pick S, Duke K, Stewart DJ, Reaume MN, Ward M, Balchin K, MacRae RM, Nelson S, Renaud J, Garvin D, Madore S, Pantarotto JR. Cancer Clinic Redesign: Opportunities for Resource Optimization. Curr Oncol 2022; 29:3983-3995. [PMID: 35735427 PMCID: PMC9222188 DOI: 10.3390/curroncol29060318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Ambulatory cancer centers face a fluctuating patient demand and deploy specialized personnel who have variable availability. This undermines operational stability through the misalignment of resources to patient needs, resulting in overscheduled clinics, budget deficits, and wait times exceeding provincial targets. We describe the deployment of a Learning Health System framework for operational improvements within the entire ambulatory center. Known methods of value stream mapping, operations research and statistical process control were applied to achieve organizational high performance that is data-informed, agile and adaptive. We transitioned from a fixed template model by an individual physician to a caseload management by disease site model that is realigned quarterly. We adapted a block schedule model for the ambulatory oncology clinic to align the regional demand for specialized services with optimized human and physical resources. We demonstrated an improved utilization of clinical space, increased weekly consistency and improved distribution of activity across the workweek. The increased value, represented as the ratio of monthly encounters per nursing worked hours, and the increased percentage of services delivered by full-time nurses were benefits realized in our cancer system. The creation of a data-informed demand capacity model enables the application of predictive analytics and business intelligence tools that will further enhance clinical responsiveness.
Collapse
Affiliation(s)
- Michael Fung-Kee-Fung
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
- Division of Gynecologic Oncology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Correspondence: (M.F.-K.-F.); (R.S.O.); Tel.: +613-737-8899 (ext. 71223) (M.F.-K.-F.)
| | - Rachel S. Ozer
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
- Correspondence: (M.F.-K.-F.); (R.S.O.); Tel.: +613-737-8899 (ext. 71223) (M.F.-K.-F.)
| | - Bill Davies
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Stephanie Pick
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Kate Duke
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - David J. Stewart
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
- Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - M. Neil Reaume
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
- Department of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Marcus Ward
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Katelyn Balchin
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Robert M. MacRae
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
- Division of Radiation Oncology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Shannon Nelson
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Julie Renaud
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Dennis Garvin
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Suzanne Madore
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
| | - Jason R. Pantarotto
- The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (B.D.); (S.P.); (K.D.); (D.J.S.); (M.N.R.); (M.W.); (K.B.); (R.M.M.); (S.N.); (J.R.); (D.G.); (S.M.); (J.R.P.)
- Division of Radiation Oncology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
13
|
Stewart DJ, Bradford JP, Batist G. Treatment Access, Health Economics, and the Wave of a Magic Wand. Curr Oncol 2022; 29:1176-1189. [PMID: 35200599 PMCID: PMC8870945 DOI: 10.3390/curroncol29020100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 11/17/2022] Open
Abstract
New drugs are expensive, in part due to excessive drug development costs. Governments are trying to reduce drug prices. This can delay access to effective agents. A country’s access to new drugs correlates with prices they agree to pay. After Health Canada approves a drug, the Canadian Agency for Drug and Technologies in Health (CADTH) assesses it. CADTH’s approval is usually contingent on it costing ≤CAD 50,000 per quality adjusted life year (QALY) gained. This value (unchanged from the 1970s) is inappropriately low. An inflation-adjusted CAD 50,000 1975 QALY should translate into a CAD 250,000 2021 QALY. CADTH’s target also does not consider that drug development costs have risen much faster than inflation or that new precision therapies may only be used in small populations. In a separate process, proposals from the Patented Medicines Price Review Board (PMPRB) would decrease initial Canadian drug prices by 20%, but prices would fall further as sales increased, with ultimate price reductions of up to 80%. PMPRB claims its proposal would not reduce drug access, but multiple analyses strongly suggest otherwise. Government price controls target the symptom (high prices), not the disease. They translate into shortages without solving the problem. CADTH and PMPRB approaches both threaten access to effective drugs.
Collapse
Affiliation(s)
- David J. Stewart
- Department of Medicine, Faculty of Medicine, The Ottawa Hospital, University of Ottawa, 501 Smyth Rd., Ottawa, ON K1H 8L6, Canada
- Life Saving Therapies Network, 173 Heath St., Ottawa, ON K1H 5E6, Canada;
- Correspondence: ; Tel.: +613-737-7700
| | | | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada;
| |
Collapse
|
14
|
Husereau D, Sullivan T, Feilotter HE, Gomes MM, Juergens R, Sheffield BS, Kassam S, Stockley TL, Jacobs P. Optimizing the delivery of genetic and advanced diagnostic testing in the province of Ontario: challenges and implications for laboratory technology assessment and management in decentralized healthcare systems. J Med Econ 2022; 25:993-1004. [PMID: 35850613 DOI: 10.1080/13696998.2022.2101807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
AIMS The Canadian province of Ontario provides full coverage for its residents (pop.14.8 M) for hospital-based diagnostic testing. Historical governance of the healthcare system and a legacy scheme of health technology assessment (HTA) and financing has led to a suboptimal approach of adopting advanced diagnostic technology (i.e. protein expression, cytogenetic, and molecular/genetic) for guiding therapeutic decisions. The aim of this research is to explore systemic barriers and provide guidance to improve patient and care provider experiences by reducing delays and inequity of access to testing, while benefitting laboratory innovators and maximizing system efficiency. MATERIALS AND METHODS A mixed-methods approach including literature review, semi-structured interviews, and a multi-stakeholder forum involving patient representatives (n = 1), laboratory leaders (n = 6), physicians (n = 5), Ministry personnel (n = 4), administrators (n = 3), extra-provincial experts, and researchers (n = 7), as well as pharmaceutical (n = 5) and diagnostic companies (n = 2). The forum considered evidence of good practices in adoption, implementation, and financing laboratory services and identified barriers as well as feasible options for improving advanced diagnostic testing in Ontario. RESULTS Overarching challenges identified included: barriers to define what is needed; need for a clear approach to adoption; and the need for more oversight and coordination. Recommendations to address these included a shift to an anticipatory system of test adoption, creating a fit-for-purpose system of health technology management that consolidates existing evaluation processes, and modernizing the governance and financing of testing so that it is managed at a care-delivery level. CONCLUSIONS The proposals for change in Ontario highlight the role that HTA, governance, and financing of health technology play along the continuum of a health technology life cycle within a healthcare system where decision-making is highly decentralized. Resource availability and capacity were not a concern - instead, solutions require higher levels of coordination and system integration along with innovative approaches to HTA.
Collapse
Affiliation(s)
- Don Husereau
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada
| | - Terrence Sullivan
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada
| | | | - Marcio M Gomes
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ontario, Canada
- Anatomical Pathologist, The Ottawa Hospital, Ottawa, Canada
| | - Rosalyn Juergens
- Division of Medical Oncology, Juravinski Cancer Centre, McMaster University, Hamilton, Canada
| | | | - Shaqil Kassam
- Stronach Regional Cancer Centre, Southlake Regional Health Centre, Newmarket, Canada
| | - Tracy L Stockley
- Division of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Philip Jacobs
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|