시장보고서
상품코드
1827233

암 프로파일링 시장 : 암 종류별, 기술별, 시료 종류별, 바이오마커 종류별, 용도별, 최종사용자별 - 세계 예측(2025-2032년)

Cancer Profiling Market by Cancer Type, Technology, Sample Type, Biomarker Type, Application, End User - Global Forecast 2025-2032

발행일: | 리서치사: 360iResearch | 페이지 정보: 영문 199 Pages | 배송안내 : 1-2일 (영업일 기준)

    
    
    




■ 보고서에 따라 최신 정보로 업데이트하여 보내드립니다. 배송일정은 문의해 주시기 바랍니다.

암 프로파일링 시장은 2032년까지 CAGR 11.73%로 288억 1,000만 달러로 성장할 것으로 예측됩니다.

주요 시장 통계
기준 연도 2024년 118억 6,000만 달러
추정 연도 2025년 131억 5,000만 달러
예측 연도 2032 288억 1,000만 달러
CAGR(%) 11.73%

암 프로파일링의 최신 기술, 임상 및 업무적 역학 관계를 간결하게 요약하여 진단 및 치료제 이해관계자의 전략적 의사결정을 지원합니다.

본 Executive Summary는 진단, 치료, 연구 이해관계자들의 전략적 계획에 도움이 될 수 있도록 암 프로파일링 기술, 임상 적용, 최종사용자 채택 동향에 대한 최신 동향을 정리한 보고서입니다. 기술 혁신과 임상적 요구의 교차점에 초점을 맞추고, 진화하는 분석 능력, 샘플 워크플로우, 바이오마커 전략이 어떻게 진단 의사결정과 중개연구의 경로를 재구성하고 있는지를 강조합니다.

차세대 시퀀싱, 분자 분석, 액체 생검 접근법의 최근 동향과 공급망 역학의 변화 및 규제 기대치를 통합하여 임상 실험실, 병원, 제약 개발 및 연구 기관에 대한 실질적인 영향을 강조합니다. 이 책은 이러한 발전을 정밀 종양학, 분자 정보에 기반한 치료법 선택, 실제 임상에서의 증거 창출과 같은 보다 광범위한 헬스케어의 요구 속에 위치시키고 있습니다. 이 책의 목적은 독자들에게 오늘날 암 프로파일링의 가장 영향력 있는 트렌드에 투자, 운영 및 임상 전략을 맞출 수 있는 간결하고 실용적인 토대를 제공하는 것입니다.

기술 중심의 분석과 최종사용자 관점의 조합을 통해 요약은 시약의 가용성, 시료 취급 기준, 데이터 해석 인프라 등 어떤 혁신 기술이 일상적인 임상적 유용성을 달성할 수 있는지를 결정하는 중요한 종속성을 밝힙니다. 이 컨텐츠는 기술 선택과 파트너십 기회를 진화하는 임상적 요구와 규제에 맞춰 조정하고자 하는 경영진, 임상 리더, 검사실 책임자에게 적합하도록 설계되었습니다.

시퀀싱, 비침습적 샘플링, 통합 바이오마커 전략의 급속한 혁신이 암 프로파일링의 임상 워크플로우와 상업적 파트너십을 재정의하는 방법

암 프로파일링은 시퀀싱의 충실도, 분석의 다중화, 임상의에게 실용적인 기간을 확대하는 샘플 진단 분석 모델의 비약적인 발전으로 인해 혁신적인 변화를 겪고 있습니다. 롱리드 및 숏리드 시퀀싱 기법의 발전이 진단 파이프라인에 통합되어 이전에는 파악하기 어려웠던 복잡한 유전체 재배열과 구조적 변이를 포착함으로써 종양 생물학 및 내성 메커니즘을 보다 세밀하게 계층화할 수 있게 되었습니다.

동시에 비침습적 샘플링과 액체 생검의 민감도 향상으로 모니터링 및 최소 잔존 병변 검출의 유용성이 확대되고, 면역조직화학 및 형광 in situ hybridization은 치료 결정을 위한 신속한 조직 기반 상황을 계속 제공하고 있습니다. 이러한 기술적 개선은 해석의 편차를 줄이고 시간을 단축하는 향상된 생물정보학 툴체인에 의해 뒷받침되며, 진단 검사실과 병원에서 폭넓게 채택될 수 있도록 돕고 있습니다.

실험실의 혁신과 함께 상호운용성, 표준화된 보고 및 크로스 플랫폼 검증을 우선시하는 모듈화된 워크플로우로 전환되고 있습니다. 이러한 움직임은 동반진단 및 치료법 선택에 직접적으로 도움이 되는 약물유전체학 지식에 대한 이해관계자들의 요구에 의해 강화되고 있으며, 분석 개발자와 제약사 간의 긴밀한 협업을 촉진하고 있습니다. 그 결과 DNA 기반, RNA 기반, 단백질 기반 바이오마커를 조합하여 다각적인 임상 정보를 제공하는 통합 진단 생태계로 패러다임이 전환되고 있습니다.

궁극적으로 이러한 변화의 누적된 효과는 전체 치료 연속체의 가치 제안을 변화시키고 있습니다. 조기에 정확한 분자 특성 분석은 진단의 오차를 줄이고, 임상시험에 대한 표적 등록을 가능하게 하며, 적응증별 치료 전략을 지원합니다. 시장 진입 기업에 대한 시사점은 분명합니다. 성공 여부는 재현 가능하고 임상적으로 실용적인 결과를 대규모로 제공할 수 있는지에 달려 있으며, 이를 뒷받침하는 것은 강력한 물류, 컴플라이언스를 준수하는 데이터 거버넌스, 공급자, 지불자, 개발자의 인센티브를 일치시키는 지속가능한 상업적 모델입니다.

2025년 관세 조치가 진단약 및 연구 분야의 암 프로파일링 관계자들의 공급망, 가격 결정력, 전략적 조달 선택에 어떤 변화를 가져올지 예측합니다.

2025년 미국의 새로운 관세 조치의 도입은 암 프로파일링 이해관계자들에게 다각적인 영향을 미칠 것이며, 공급망, 조달 전략, 국경 간 협력에 영향을 미칠 것입니다. 시퀀싱 장비 및 어세이 키트 제조업체의 경우, 일부 부품 및 완제품에 대한 수입 관세 인상으로 인해 상륙 비용이 상승할 수 있으며, 공급업체는 가격 책정, 조달 및 재고를 재검토해야 할 것으로 보입니다. 이러한 움직임은 진단 실험실과 병원에 시약 구매 주기를 재평가하고, 임상 처리량을 손상시키지 않으면서도 고부가가치 분석에 대한 접근성을 유지할 수 있는 계약을 협상하도록 압력을 가할 것입니다.

관세는 직접적인 비용 효과뿐만 아니라 공급업체 다변화, 주요 제조 공정의 니어쇼어링, 리드타임 변동성을 완화하기 위한 지역적 유통 거점 중시 등 전략적 대응을 촉진할 수 있습니다. 이러한 적응은 중기적으로 회복력을 높일 수 있지만, 일시적으로 서비스 제공에 지장을 초래할 수 있는 선행 투자 및 운영 변경이 필요합니다. 이와 함께 수입된 특수 시약과 장비에 의존하는 연구기관과 제약회사는 프로젝트 지연과 연구비용 증가에 직면하여 바이오마커 검증과 동반진단약 개발 일정에 영향을 미칠 수 있습니다.

관세 주도의 가격 조정은 롱 리드 시퀀스나 하이 컨텐츠 플랫폼과 같은 첨단 기술 채택에도 영향을 미칩니다. 자원이 제한된 환경의 의료 서비스 제공자는 면역조직화학이나 중합효소 연쇄 반응과 같은 저비용의 확립된 방법을 우선시하여 고비용이지만 임상적으로 유용한 기술의 보급을 지연시킬 수 있습니다. 반대로, 지불자와 의료 시스템은 전체 지출을 줄이면서 종합적인 프로파일링에 대한 접근성을 유지하기 위해 우수 센터 내에서 검사의 통합을 가속화할 수 있습니다.

규제 및 정책적 측면에서, 관세는 생명공학 공급망의 전략적 중요성에 대한 산업계와 정부 간의 대화를 촉진하고, 국내 제조, 민관 파트너십, 현지 역량에 대한 집중적인 투자에 대한 인센티브를 가속화할 수 있습니다. 이러한 정책적 대응은 장기적으로 취약성을 줄일 수 있지만, 동시에 경쟁 역학과 제조 노하우의 세계 분포를 형성하게 될 것입니다. 관세 환경을 종합하면, 고품질 암 프로파일링 서비스의 연속성을 보장하기 위해서는 공급망의 투명성, 유연한 조달 전략, 적극적인 이해관계자 참여가 중요하다는 것을 알 수 있습니다.

부문 수준의 분석을 통해 암 유형, 기술 선택, 샘플 매트릭스, 바이오마커 클래스, 애플리케이션 목적 및 최종사용자 요구사항이 임상 및 상업적 우선순위를 결정하는 방법을 파악할 수 있습니다.

세분화를 통해 얻은 인사이트는 임상적 유용성, 기술 성숙도, 최종사용자의 요구가 암 프로파일링 상황 전반에 걸쳐 기회와 위험을 창출하는 수렴점을 밝혀냅니다. 혈액학 워크플로우에서는 백혈병, 림프종, 다발성 골수종에서 특이적 융합 사건, 클론 동태, 최소 잔존 병변의 검출에 중점을 두는 반면, 고형 종양 프로파일링에서는 유방암, 대장암, 폐암, 전립선암에서 실용적인 돌연변이 및 종양 불균일성에 중점을 둡니다. 이러한 다양한 임상 목표는 검사실 업무에서 분석법 선택, 시료 취급 및 보고의 세분화에 영향을 미칩니다.

목차

제1장 서론

제2장 분석 방법

제3장 주요 요약

제4장 시장 개요

제5장 시장 인사이트

제6장 미국 관세의 누적 영향 2025

제7장 AI의 누적 영향 2025

제8장 암 프로파일링 시장 : 암 종류별

  • 조혈 악성 종양
    • 백혈병
    • 림프종
    • 다발성 골수종
  • 고형 종양
    • 유방암
    • 대장암
    • 폐암
    • 전립선암

제9장 암 프로파일링 시장 : 기술별

  • FISH(Fluorescent In Situ Hybridization)
  • 면역조직화학
  • 마이크로어레이
  • 차세대 시퀀싱
    • 롱리드 시퀀싱
    • 쇼트 리드 시퀀싱
  • 중합효소 연쇄 반응(PCR)

제10장 암 프로파일링 시장 : 시료 종류별

  • 혈액
    • 혈장
    • 혈청
    • 전혈
  • 타액
  • 소변

제11장 암 프로파일링 시장 : 바이오마커 종류별

  • DNA 기반
  • 단백질 기반
  • RNA 기반

제12장 암 프로파일링 시장 : 용도별

  • 진단
  • 모니터링
    • 액체 생검
    • 미세잔존질환
  • 예후
  • 연구
  • 치료법 선택
    • 동반진단
    • 약물유전체학

제13장 암 프로파일링 시장 : 최종사용자별

  • 진단 검사실
  • 병원
  • 제약 기업
  • 연구기관

제14장 암 프로파일링 시장 : 지역별

  • 아메리카
    • 북미
    • 라틴아메리카
  • 유럽, 중동 및 아프리카
    • 유럽
    • 중동
    • 아프리카
  • 아시아태평양

제15장 암 프로파일링 시장 : 그룹별

  • ASEAN
  • GCC
  • EU
  • BRICS
  • G7
  • NATO

제16장 암 프로파일링 시장 : 국가별

  • 미국
  • 캐나다
  • 멕시코
  • 브라질
  • 영국
  • 독일
  • 프랑스
  • 러시아
  • 이탈리아
  • 스페인
  • 중국
  • 인도
  • 일본
  • 호주
  • 한국

제17장 경쟁 구도

  • 시장 점유율 분석(2024년)
  • FPNV 포지셔닝 매트릭스(2024년)
  • 경쟁 분석
    • F. Hoffmann-La Roche AG
    • Thermo Fisher Scientific Inc.
    • 4basecare Onco Solutions Private Limited
    • ACT Genomics Co., Ltd. by Prenetics Global Limited
    • Agendia, Inc.
    • Agilent Technologies, Inc.
    • BostonGene Corporation
    • Caris Life Sciences
    • Exact Sciences Corporation
    • GENINUS Inc.
    • Genomic Life
    • GenScript Biotech Corporation
    • Guardant Health, Inc.
    • Hologic, Inc.
    • HTG Molecular Diagnostics, Inc.
    • Illumina, Inc.
    • IMBdx, Inc.
    • Laboratory Corporation of America Holdings
    • Lucence Health, Inc.
    • Merck KGaA
    • NanoString Technologies, Inc.
    • Neogenomics, Inc.
    • Nonacus Limited
    • OncoDNA S.A.
    • Oncompass Medicine Hungary Kft.
    • Paragon Genomics, Inc.
    • Personalis, Inc.
    • Perthera, Inc.
    • Qiagen N.V.
    • Renovaro Biosciences Inc.
    • Strand Life Sciences
    • Sysmex Corporation
    • Takara Bio Inc.
    • Tempus Labs Inc.
KSM 25.10.16

The Cancer Profiling Market is projected to grow by USD 28.81 billion at a CAGR of 11.73% by 2032.

KEY MARKET STATISTICS
Base Year [2024] USD 11.86 billion
Estimated Year [2025] USD 13.15 billion
Forecast Year [2032] USD 28.81 billion
CAGR (%) 11.73%

A concise synthesis of recent technological, clinical, and operational dynamics in cancer profiling that frames strategic decisions for diagnostics and therapeutic stakeholders

This executive summary synthesizes contemporary developments in cancer profiling technologies, clinical applications, and end-user adoption trends to inform strategic planning for stakeholders across diagnostics, therapeutics, and research. It focuses on the intersection of technological innovation and clinical need, highlighting how evolving assay capabilities, sample workflows, and biomarker strategies are reshaping diagnostic decision-making and translational research pathways.

The narrative emphasizes practical implications for clinical laboratories, hospitals, pharmaceutical developers, and research institutions by integrating recent advances in next-generation sequencing, molecular assays, and liquid biopsy approaches with shifting supply chain dynamics and regulatory expectations. It situates these developments within broader healthcare imperatives such as precision oncology, molecularly informed therapy selection, and real-world evidence generation. The intent is to provide readers with a concise, actionable foundation for aligning investment, operational, and clinical strategies with the most impactful trends in cancer profiling today.

Through a combination of technology-focused analysis and end-user perspectives, the summary also identifies critical dependencies-such as reagent availability, sample handling standards, and data interpretation infrastructures-that will determine which innovations achieve routine clinical utility. The content is designed to be relevant for executives, clinical leaders, and laboratory directors seeking to weigh technology choices and partnership opportunities against evolving clinical demands and regulatory pathways.

How rapid innovations in sequencing, noninvasive sampling, and integrated biomarker strategies are redefining clinical workflows and commercial partnerships in cancer profiling

Cancer profiling is undergoing transformative shifts driven by leaps in sequencing fidelity, assay multiplexing, and sample-agnostic analytic models that expand the actionable window for clinicians. Advances in long-read and short-read sequencing modalities are being integrated into diagnostic pipelines to capture complex genomic rearrangements and structural variants that were previously elusive, enabling finer stratification of tumor biology and resistance mechanisms.

Concurrently, improvements in noninvasive sampling and liquid biopsy sensitivity are extending the utility of monitoring and minimal residual disease detection, while immunohistochemistry and fluorescence in situ hybridization continue to provide rapid, tissue-based context for therapeutic decisions. These technological refinements are supported by enhanced bioinformatics toolchains that reduce interpretive variability and accelerate turnaround times, fostering broader adoption among diagnostic laboratories and hospitals.

Parallel to laboratory innovations, the landscape is shifting toward modular workflows that prioritize interoperability, standardized reporting, and cross-platform validation. This movement is reinforced by stakeholders' demand for companion diagnostics and pharmacogenomic insights that directly inform therapy selection, driving closer collaboration between assay developers and pharmaceutical companies. As a result, the paradigm is moving from isolated tests toward integrated diagnostic ecosystems that combine DNA-based, RNA-based, and protein-based biomarkers to deliver multidimensional clinical intelligence.

Finally, the cumulative effect of these shifts is altering value propositions across the care continuum: early and accurate molecular characterization reduces diagnostic odysseys, enables targeted enrollment into clinical trials, and supports adaptive treatment strategies. The implication for market participants is clear: success will depend on delivering reproducible, clinically actionable results at scale, supported by robust logistics, compliant data governance, and sustainable commercial models that align incentives across providers, payers, and developers.

Anticipating how 2025 tariff measures reshape supply chains, pricing dynamics, and strategic sourcing choices for cancer profiling stakeholders across diagnostics and research

The introduction of new tariff measures in the United States in 2025 presents a multifaceted set of implications for cancer profiling stakeholders, affecting supply chains, procurement strategies, and cross-border collaboration. For manufacturers of sequencing instruments and assay kits, increased import duties on select components and finished goods can elevate landed costs, prompting suppliers to reassess pricing, sourcing, and inventory practices. This dynamic will exert pressure on diagnostic laboratories and hospitals to reevaluate reagent purchasing cycles and to negotiate contracts that preserve access to high-value assays without compromising clinical throughput.

Beyond direct cost effects, tariffs can catalyze strategic responses such as supplier diversification, nearshoring of critical manufacturing steps, and greater emphasis on regional distribution hubs to mitigate lead-time volatility. These adaptations may improve resilience in the medium term but require upfront capital and operational changes that can temporarily disrupt service delivery. In parallel, research institutes and pharmaceutical companies that depend on imported specialized reagents and instrumentation may face project delays or increased study costs, influencing timelines for biomarker validation and companion diagnostic development.

Tariff-driven price adjustments also have downstream implications for adoption of advanced technologies such as long-read sequencing or high-content platforms. Providers in resource-constrained settings may prioritize established, lower-cost methodologies like immunohistochemistry and polymerase chain reaction, slowing diffusion of higher-cost but clinically informative technologies. Conversely, payers and health systems may accelerate consolidation of testing within centers of excellence to preserve access to comprehensive profiling while containing overall expenditure.

On the regulatory and policy front, tariffs can prompt dialogue between industry and government about strategic importance of biotech supply chains, potentially accelerating incentives for domestic manufacturing, public-private partnerships, and targeted investments in local capacity. These policy responses may reduce long-term vulnerability but will also shape competitive dynamics and the distribution of manufacturing expertise globally. Collectively, the tariff environment underscores the importance of supply chain transparency, flexible sourcing strategies, and proactive stakeholder engagement to ensure continuity of high-quality cancer profiling services.

Segment-level analysis revealing how cancer type, technology choice, sample matrix, biomarker class, application purpose, and end-user requirements jointly determine clinical and commercial priorities

Insights derived from segmentation illuminate where clinical utility, technology maturity, and end-user needs converge to create opportunity and risk across the cancer profiling landscape. Based on cancer type, differences between hematologic malignancies and solid tumors drive distinct testing priorities: hematologic workflows emphasize detection of specific fusion events, clonal dynamics, and minimal residual disease across leukemia, lymphoma, and multiple myeloma, while solid tumor profiling concentrates on actionable mutations and tumor heterogeneity within breast, colorectal, lung, and prostate cancers. These divergent clinical goals inform assay selection, sample handling, and reporting granularity in laboratory operations.

Based on technology, the diagnostic toolkit spans fluorescence in situ hybridization, immunohistochemistry, microarray platforms, next-generation sequencing, and polymerase chain reaction. Within next-generation sequencing, the emergence of both long-read sequencing and short-read sequencing enables complementary interrogation of genomic complexity: short-read approaches provide high-throughput mutation detection and cost-efficiency, while long-read methods resolve structural variants and repetitive regions that have clinical relevance for specific tumor types. The choice of technology is shaped by clinical indication, turnaround time requirements, and the analytic sensitivity necessary for monitoring and early detection.

Based on sample type, blood, saliva, and urine each offer unique advantages and constraints. Blood-derived analyses-including plasma, serum, and whole blood-support both tumor genotyping and longitudinal monitoring via circulating tumor DNA, whereas saliva and urine present noninvasive alternatives for certain tumor types and patient populations. Sample matrices influence pre-analytical workflows, cold chain logistics, and interpretation frameworks, requiring laboratories to standardize collection protocols to preserve assay performance.

Based on biomarker type, DNA-based, protein-based, and RNA-based markers provide orthogonal insights that enhance diagnostic confidence and therapeutic alignment. DNA analyses excel at identifying somatic and germline variants; RNA-based assays reveal gene expression patterns and fusion transcripts; and protein-based measurements capture phenotypic states that can predict drug response. Integrating these biomarker classes into composite reports increases clinical actionability but demands harmonized validation and cross-platform comparability.

Based on application, diagnosis, monitoring, prognosis, research, and therapy selection define the primary use cases for profiling technologies. Monitoring use cases such as liquid biopsy and minimal residual disease require assays with exceptional sensitivity and reproducibility, while therapy selection relies on companion diagnostics and pharmacogenomics to match patients to targeted therapies and dosing strategies. Each application imposes different demands on assay design, regulatory documentation, and evidence generation.

Based on end user, diagnostic laboratories, hospitals, pharmaceutical companies, and research institutes exhibit distinct operational constraints and value drivers. Diagnostic laboratories prioritize throughput, reproducibility, and reimbursement pathways; hospitals focus on integration with clinical workflows and electronic health records; pharmaceutical companies seek robust biomarker validation and companion diagnostic alignment for drug development; and research institutes emphasize exploratory analyses and novel target discovery. Recognizing these user-specific imperatives is essential for tailoring product design, service models, and commercialization strategies.

A regional perspective on adoption pathways, regulatory variability, and infrastructure requirements that influence equitable access and scale-up of cancer profiling solutions

Regional dynamics play a decisive role in shaping access, adoption rates, and strategic priorities for cancer profiling across the globe. In the Americas, investments in precision oncology infrastructure, widespread adoption of next-generation sequencing, and integrated care networks support rapid clinical translation of novel assays, while market participants must navigate reimbursement complexity and heterogeneous laboratory accreditation landscapes. This region often leads in companion diagnostic collaborations between device makers and pharmaceutical sponsors, producing rich pipelines for implementation studies and real-world evidence generation.

In Europe, Middle East & Africa, heterogeneous regulatory frameworks and variable healthcare financing models create a patchwork of adoption scenarios that favor regionally tailored strategies. High-capacity centers in Western Europe act as hubs for advanced diagnostics and multicenter studies, whereas other subregions prioritize cost-effective assays and scalable workflows that can operate in constrained settings. Cross-border collaborations within Europe are increasingly important for harmonizing standards, enabling data sharing, and accelerating multicountry trials that validate biomarker utility across diverse populations.

In Asia-Pacific, rapid expansion of laboratory capacity, increasing public and private investment in genomics, and large patient populations present substantial opportunities for scaling both diagnostic services and biomarker-driven clinical research. Nevertheless, varying levels of regulatory maturity and differences in reimbursement create a landscape where local partnerships, technology transfer, and capacity building are critical to achieving broad access. Across all regions, supply chain considerations, workforce training, and data governance frameworks will determine the pace and equity of clinical implementation, making regionally informed strategies essential for sustained impact.

How strategic specialization, integrated service models, and collaborative companion diagnostic partnerships define competitive advantage and enable scalable clinical adoption

Leading companies and research organizations are differentiating through specialization, vertically integrated offerings, and collaborative models that combine assay development with informatics and clinical support. Some firms concentrate on platform excellence-delivering validated instruments, consumables, and standardized workflows that enable high-throughput diagnostics-while others focus on niche clinical applications such as minimal residual disease detection or complex structural variant resolution. Strategic partnerships between technology providers and pharmaceutical developers are increasingly common, aligning companion diagnostic development with therapeutic pipelines to streamline regulatory approval and market access.

Competitive dynamics hinge on the ability to demonstrate analytical validity, clinical utility, and operational scalability. Organizations that invest in rigorous multicenter validation studies and interoperable reporting standards gain credibility with laboratories and payers. In addition, companies that provide end-to-end solutions-including sample logistics, bioinformatics, and clinician-facing interpretation-are better positioned to capture value in hospital and laboratory settings that prefer turnkey implementations.

Mergers, acquisitions, and licensing agreements continue to shape the landscape as firms seek to broaden their portfolios and secure access to complementary technologies and regional distribution networks. Simultaneously, a growing cohort of specialized service providers supports research institutes and pharmaceutical companies with bespoke assay development, biomarker validation, and translational analytics. This ecosystem of platform vendors, service providers, and clinical adopters is creating multiple pathways for innovation diffusion, each with distinct implications for pricing, reimbursement negotiation, and long-term sustainability.

Actionable strategic priorities for leaders to build supply resilience, clinical validation pathways, and interoperable services that accelerate uptake and stewardship of profiling solutions

Industry leaders should prioritize investments that reinforce resilience, clinical relevance, and seamless integration into care pathways. Strengthening supply chain transparency and sourcing flexibility will mitigate tariff and logistics risks, while targeted investments in regional manufacturing or distribution can shorten lead times and stabilize access to critical consumables. Concurrently, organizations should invest in cross-platform validation studies that demonstrate concordance between technologies and support interoperability of clinical reports across electronic health systems.

From a product and clinical strategy standpoint, companies should align assay development with high-impact clinical use cases such as therapy selection and minimal residual disease monitoring, producing evidence packages that address payer and clinician requirements. Collaboration with pharmaceutical sponsors for co-development of companion diagnostics and with academic centers for prospective validation studies will accelerate clinical uptake. Equally important is building interpretive services and decision-support tools that translate complex molecular results into actionable clinical recommendations, reducing adoption friction among clinicians.

Operationally, stakeholders must expand workforce training and establish robust quality systems to sustain reproducibility as testing volumes grow. Leaders should also engage proactively with regulators and payers to shape evidence expectations and reimbursement frameworks, emphasizing real-world performance and patient-centric outcomes. Finally, adopting flexible commercial models-ranging from centralized reference testing to reagent-as-a-service-can help organizations tailor offerings to diverse end-user needs and create multiple revenue streams while maintaining patient access.

A transparent and practitioner-focused methodology combining expert engagements, technical literature synthesis, and cross-validation against laboratory standards to ensure actionable insights

The research methodology underpinning this executive summary combined primary and secondary sources, expert interviews, and structured synthesis to produce a balanced assessment of technological, clinical, and commercial trends. Primary inputs included conversations with laboratory directors, clinical investigators, and industry executives to capture operational realities, evidence requirements, and adoption barriers. These qualitative engagements were supplemented by careful review of peer-reviewed literature, regulatory guidance, and technical white papers to ground technological descriptions and clinical use-case delineations.

Analytic approaches emphasized triangulation: cross-referencing expert perspectives with published validation studies and technical specifications to identify consistent patterns of performance and adoption. Particular attention was paid to assay sensitivity, specificity, and workflow compatibility, as well as to logistics such as sample stability and cold chain dependencies. Where possible, methodological choices were benchmarked against established laboratory standards and consensus recommendations to ensure practical relevance.

Limitations of the methodology include potential bias inherent in selective expert sampling and variability in publicly available technical documentation. To mitigate these constraints, the study prioritized sources with direct operational experience and sought corroboration across multiple stakeholder types. The result is a defensible, practitioner-oriented set of insights designed to support strategic decision-making without relying on proprietary market estimates.

Final synthesis underscoring the critical interplay of technology, evidence, and operational resilience that will determine which profiling approaches achieve sustained clinical impact

In conclusion, cancer profiling is at an inflection point where technical advances, regulatory evolution, and shifting supply chain considerations converge to create both opportunities and operational challenges. The most successful organizations will be those that integrate robust assay technology with validated workflows, responsive supply strategies, and clinician-centered reporting that directly support therapeutic decision-making. Emphasizing cross-platform validation and evidence generation will be critical for translating analytical capabilities into routine clinical use.

Moreover, regional heterogeneity and policy dynamics will require adaptive commercial models and sustained stakeholder engagement. By focusing on resilience, clinical alignment, and partnerships that bridge technological and therapeutic development, leaders can accelerate the adoption of high-impact profiling solutions while safeguarding continuity of care. The insights presented here aim to equip senior decision-makers with the context and priorities needed to navigate this evolving landscape and to identify the investments that will yield enduring clinical and commercial value.

Table of Contents

1. Preface

  • 1.1. Objectives of the Study
  • 1.2. Market Segmentation & Coverage
  • 1.3. Years Considered for the Study
  • 1.4. Currency & Pricing
  • 1.5. Language
  • 1.6. Stakeholders

2. Research Methodology

3. Executive Summary

4. Market Overview

5. Market Insights

  • 5.1. Adoption of circulating tumor DNA liquid biopsy assays for real-time treatment monitoring in oncology
  • 5.2. Implementation of multiomic profiling platforms combining genomics proteomics and metabolomics data for comprehensive tumor characterization
  • 5.3. Regulatory approval trends for next-generation sequencing-based companion diagnostics in targeted cancer therapy selection
  • 5.4. Expansion of artificial intelligence and machine learning algorithms to interpret complex cancer profiling datasets and predict patient outcomes
  • 5.5. Development of cost-effective point-of-care molecular profiling devices for early cancer detection in low-resource settings
  • 5.6. Emergence of epigenetic biomarker panels to stratify patient response to immunotherapy across solid tumor indications
  • 5.7. Partnerships between biopharma and diagnostic companies to co-develop liquid biopsy assays for minimal residual disease monitoring
  • 5.8. Incorporation of spatial transcriptomics into clinical workflows to map tumor microenvironment heterogeneity with high resolution

6. Cumulative Impact of United States Tariffs 2025

7. Cumulative Impact of Artificial Intelligence 2025

8. Cancer Profiling Market, by Cancer Type

  • 8.1. Hematologic Malignancies
    • 8.1.1. Leukemia
    • 8.1.2. Lymphoma
    • 8.1.3. Multiple Myeloma
  • 8.2. Solid Tumors
    • 8.2.1. Breast
    • 8.2.2. Colorectal
    • 8.2.3. Lung
    • 8.2.4. Prostate

9. Cancer Profiling Market, by Technology

  • 9.1. Fluorescence In Situ Hybridization
  • 9.2. Immunohistochemistry
  • 9.3. Microarray
  • 9.4. Next Generation Sequencing
    • 9.4.1. Long Read Sequencing
    • 9.4.2. Short Read Sequencing
  • 9.5. Polymerase Chain Reaction

10. Cancer Profiling Market, by Sample Type

  • 10.1. Blood
    • 10.1.1. Plasma
    • 10.1.2. Serum
    • 10.1.3. Whole Blood
  • 10.2. Saliva
  • 10.3. Urine

11. Cancer Profiling Market, by Biomarker Type

  • 11.1. DNA-Based
  • 11.2. Protein-Based
  • 11.3. RNA-Based

12. Cancer Profiling Market, by Application

  • 12.1. Diagnosis
  • 12.2. Monitoring
    • 12.2.1. Liquid Biopsy
    • 12.2.2. Minimal Residual Disease
  • 12.3. Prognosis
  • 12.4. Research
  • 12.5. Therapy Selection
    • 12.5.1. Companion Diagnostics
    • 12.5.2. Pharmacogenomics

13. Cancer Profiling Market, by End User

  • 13.1. Diagnostic Laboratories
  • 13.2. Hospitals
  • 13.3. Pharmaceutical Companies
  • 13.4. Research Institutes

14. Cancer Profiling Market, by Region

  • 14.1. Americas
    • 14.1.1. North America
    • 14.1.2. Latin America
  • 14.2. Europe, Middle East & Africa
    • 14.2.1. Europe
    • 14.2.2. Middle East
    • 14.2.3. Africa
  • 14.3. Asia-Pacific

15. Cancer Profiling Market, by Group

  • 15.1. ASEAN
  • 15.2. GCC
  • 15.3. European Union
  • 15.4. BRICS
  • 15.5. G7
  • 15.6. NATO

16. Cancer Profiling Market, by Country

  • 16.1. United States
  • 16.2. Canada
  • 16.3. Mexico
  • 16.4. Brazil
  • 16.5. United Kingdom
  • 16.6. Germany
  • 16.7. France
  • 16.8. Russia
  • 16.9. Italy
  • 16.10. Spain
  • 16.11. China
  • 16.12. India
  • 16.13. Japan
  • 16.14. Australia
  • 16.15. South Korea

17. Competitive Landscape

  • 17.1. Market Share Analysis, 2024
  • 17.2. FPNV Positioning Matrix, 2024
  • 17.3. Competitive Analysis
    • 17.3.1. F. Hoffmann-La Roche AG
    • 17.3.2. Thermo Fisher Scientific Inc.
    • 17.3.3. 4basecare Onco Solutions Private Limited
    • 17.3.4. ACT Genomics Co., Ltd. by Prenetics Global Limited
    • 17.3.5. Agendia, Inc.
    • 17.3.6. Agilent Technologies, Inc.
    • 17.3.7. BostonGene Corporation
    • 17.3.8. Caris Life Sciences
    • 17.3.9. Exact Sciences Corporation
    • 17.3.10. GENINUS Inc.
    • 17.3.11. Genomic Life
    • 17.3.12. GenScript Biotech Corporation
    • 17.3.13. Guardant Health, Inc.
    • 17.3.14. Hologic, Inc.
    • 17.3.15. HTG Molecular Diagnostics, Inc.
    • 17.3.16. Illumina, Inc.
    • 17.3.17. IMBdx, Inc.
    • 17.3.18. Laboratory Corporation of America Holdings
    • 17.3.19. Lucence Health, Inc.
    • 17.3.20. Merck KGaA
    • 17.3.21. NanoString Technologies, Inc.
    • 17.3.22. Neogenomics, Inc.
    • 17.3.23. Nonacus Limited
    • 17.3.24. OncoDNA S.A.
    • 17.3.25. Oncompass Medicine Hungary Kft.
    • 17.3.26. Paragon Genomics, Inc.
    • 17.3.27. Personalis, Inc.
    • 17.3.28. Perthera, Inc.
    • 17.3.29. Qiagen N.V.
    • 17.3.30. Renovaro Biosciences Inc.
    • 17.3.31. Strand Life Sciences
    • 17.3.32. Sysmex Corporation
    • 17.3.33. Takara Bio Inc.
    • 17.3.34. Tempus Labs Inc.
샘플 요청 목록
0 건의 상품을 선택 중
목록 보기
전체삭제