
Latest Developments in Precision Medicine
Explore recent advances—from diagnostic technologies to novel biomarkers aiming to transform precision medicine.
Jump to:
Digital Pathology Adoption Is Growing Due to
a Number of Advantages
Computerized workflows and digitized image analysis are helping healthcare providers interpret complex data, creating personalized solutions for patients.1–3
Digital pathology may provide several benefits in precision medicine, including:

- Increased diagnostic accuracy and predictivity compared to traditional pathology methods4

- Paperless transmission of digital slides to and from members of the multidisciplinary team (MDT), reducing chances of misidentification or transposition errors5

- Optimization of workflow through quick and efficient tracking5
- Transfer of slides and diagnostic information from laboratory to pathologist5

By using digital pathology, you can reduce persistent diagnostic testing challenges and provide faster delivery of results.5
MDT, multidisciplinary team.
Combine Digital Pathology With Computational Pathology for Enhanced Insights
When used with digital pathology, computational pathology extracts and analyzes complex data sets of pathology images to potentially improve workflow efficiencies and diagnostic quality.6,7 Combine digital pathology with genomic, clinical, and other data for novel diagnostic insights.
Technology | Benefit in practice |
---|---|
Incorporates multiple channels of data (e.g., demographics, digital pathology, multi-omics, lab results) into mathematic models.6,8 |
|
With whole slide imaging, deep learning models in computational pathology are trained to detect malignancies and classify histology subtypes.8 |
|
Integrating AI-driven pathology image analysis with radiomics to bridge medical imaging and precision medicine.8 |
|
AI, artificial intelligence; FDA, US Food and Drug Administration; HER2, human epidermal growth factor receptor 2.
Diagnostic Advancements Are Improving Cancer
Diagnosis and Care
Cancer diagnosis is often made at advanced stages where treatment choices are limited. Molecular biomarkers
have spearheaded advancements in early cancer detection, with emerging methods geared towards further
improving detection and monitoring.15

When modelling for maximum benefit,
the use of MCED may result
in a relative
26%18,a
mortality reduction
Detect more cancers at treatable stages with multi-cancer early detection (MCED) screening
MCED is an investigational blood-based analysis that can test for a variety of cancers at once, rather than just one at a time.16,17
Many MCED tests are under investigation to detect cancer signals in DNA sequences, RNA sequences, DNA methylation, DNA fragmentation, protein levels, or antibodies in the blood. Currently, no MCED test is recommended in professional guidelines.16.Eventually, MCED’s use of biomarker testing as a screening tool may help find cancers at earlier stages.16,18

MRD Monitoring
Diagnosis
Treatment
Post-treatment
ctDNA
Time from treatment
Monitor minimal residual disease (MRD) to inform future treatment
Knowing the MRDÂ status of a patient in terms of the presence or absence of a very small number of tumor cells that have survived treatment19 may help you decide whether to continue treatment to improve outcomes or de-escalate treatment to reduce toxicities.20
MRD is usually assessed using traces of circulating tumor DNA (ctDNA) in the blood. Other MRD signals have also been explored, such as circulating tumor cells (CTCs) and non-coding RNA.21 While MRD is more established in blood cancers,22,23 it is also being investigated in solid tumors, including lung,24,25 breast,26,27 ovarian,28 and prostate cancers.29
Footnotes
aUnder maximum benefit, 267 (68%) of those individuals would be intercepted before usual care. Applying the hazard reduction from stage at interception when compared with original stage at diagnosis, only 163 of those individuals would die within 5 years of the original diagnosis date, a 39% reduction in mortality for an absolute reduction of 104 deaths per 100,000, resulting in a relative all-cancer mortality reduction of 26%.18
CTC, circulating tumor cell; ctDNA, circulating tumor DNA; DNA, deoxyribonucleic acid; MCED, multi-cancer early detection; MRD, minimal residual disease; RNA, ribonucleic acid.
Novel Biomarkers Reveal Potential
Treatment Opportunities
An increasing armamentarium of biomarkers can help support cancer diagnosis and treatment at the earliest stage possible. Novel biomarkers can help identify new patient subsets, complement established biomarkers, and provide MDTs with increasingly informed and efficient options in cancer treatment.30
The Association of Molecular Pathology (AMP) offers a wealth of resources, including webinars on evolving biomarkers.
View Association of Molecular Pathology resources
Novel Biomarkers May Have Prognostic and Predictive Value to Patients With Metastatic NSCLC
Emerging data point to STK11, KEAP1, and/or SMARCA4Â as novel biomarkers that may help guide clinical management of patients with metastatic NSCLC.31 Small, retrospective, exploratory subgroup analyses in NSCLC suggest that:
Potentially Prognostic
Inactivation of these genes may negatively impact disease outcomes in patients with NSCLC32–35
- STK11 inactivation
- KEAP1 inactivation
- SMARCA4 inactivation
Potentially Predictive
These genes are potentially associated with poorer response to immunotherapy regimens vs wild-type33,34,36
- STK11 mutationsa
- KEAP1 mutationsb
- SMARCA4 mutation + KRAS co-mutation (36% co-occurrence, compared with KRAS mutations alone)c
This gene is potentially associated with positive response to immunotherapy regimens vs wild type32,37
- SMARCA4 mutationc
Prevalence of STK11, KEAP1, and SMARCA4 in advanced/metastatic NSCLC38,39,d,e
Gene | Average frequency (%)f | Frequency range (%) |
---|---|---|
STK11 | 17 | 16–20 |
KEAP1 | 17 | 16–20 |
SMARCA4 | 10 | N/A |
- KRAS-mutant NSCLC represents a large and highly heterogeneous patient subgroup (~30% of NSCLC) with a poorer prognosis and a high comutation rate that can complicate treatment planning37,40–42Â
- KRAS mutations frequently co-occur with STK11, KEAP1, and/or SMARCA4 mutations32,35,38Â
- These comutated subgroups may be associated with suboptimal outcomes to IO regimens32,34–36,43,g
- Additional investigation is needed to further characterize novel biomarkers of interest (including mutations in STK11, KEAP1, and SMARCA4) that have the potential to aid in patient selection for 1L immunotherapy regimens in patients with NSCLC
- To help better inform treatment planning for metastatic NSCLC patients who are eligible for 1L immunotherapy, novel prognostic and predictive biomarkers are an evolving area of research.31
Footnotes
aBased on 2 retrospective studies of nonsquamous NSCLC patients: a pooled analysis of 32 patients from 2 trials who had an STK11 mutation and had received a single-agent PD-(L)1 inhibitor,33 and a cohort of 66 PD-L1–positive (≥1%) patients with STK11 mutations (irrespective of KRAS status) who received single-agent PD-(L)1 inhibitor therapy or combination PD-(L)1 inhibitor and CTLA-4 inhibitor therapy.36 Patient staging and previous lines of therapy were not specified in these analyses.33,36 bBased on 2 retrospective studies of NSCLC patients: a pooled analysis of 45 patients with nonsquamous NSCLC from 2 trials who had a KEAP1 mutation and had received a single-agent PD-(L)1 inhibitor,33 and an analysis of 2 cohorts of patients with lung adenocarcinoma and a KEAP1 mutation. Cohort 1, the blood-based NGS cohort, analyzed 304 patients from 2 studies who received single-agent PD-(L)1 inhibitor therapy. Cohort 2, the tissue-based NGS cohort of 343 patients derived from a cohort and 2 studies did not specify the immunotherapy regimen.34 Patient staging and previous lines of therapy were not specified in these analyses.33,34 cBased on 2 retrospective studies of NSCLC patients: an analysis of 2 separate cohorts of 77 and 18 patients with KRAS mutations with or without SMARCA4 mutations and who had received immunotherapy,37 and a cohort of 86 patients with SMARCA4 mutations across all histologies and of any stage who had received an immunotherapy regimen.32 Patient staging was not specified in the first analysis and the immunotherapy regimen and previous lines of therapy were not specified in either analysis.32,37 dThere is prevalent co-occurrence of these 3 mutations in NSCLC. A retrospective analysis of real-world data in patients with Stages IIIB, IIIC, IVA, and IVB nonsquamous NSCLC found that tumors carrying either STK11 and/or KEAP1 mutations occurred in 30% of patients (n=2276).38 In a single-institution retrospective analysis of patients with any stage of NSCLC, SMARCA4 mutations co-occurred with KEAP1 in 41% of SMARCA4-mutant tumors and with STK11 in 39% of SMARCA4-mutant tumors (n=407).32 eBased on 2 retrospective analyses of real-world data from NSCLC patients: an analysis of 2276 patients from a database with Stage IIIB, IIIC, IVA, or IVB nonsquamous NSCLC classified by 1L treatment across 5 treatment classes (STK11 and KEAP1 only),38 and an analysis of 12,934 patients from a database with advanced NSCLC (STK11, KEAP1, and SMARCA4).39 Disease stage and histology were not specified in the second analysis.39 fAverage frequencies were calculated as weighted averages; weighting was based on the total number of participants in each study. Frequency for SMARCA4 is not an average, but based on Liu et al.39 Please see additional details in footnote e. gBased on 4 retrospective analyses: a retrospective analysis of 532 patients with advanced NSCLC (stage and histology not specified) who had received at least one dose of a PD-L(1) inhibitor alone or in combination with a CTLA-4 inhibitor,43 and a retrospective subgroup analysis of 1202 patients with Stage IV metastatic nonsquamous NSCLC who had received a PD-L1 inhibitor + chemotherapy, a VEGF inhibitor + chemotherapy, or a PD-L1 inhibitor + a VEGF inhibitor + chemotherapy as 1L therapy.35 Please see footnote a for the details of reference 36, and footnote b for the details of reference 34.
1L, first-line; AMP, Association of Molecular Pathology; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; KEAP1, kelch-like ECH-associated protein 1; KRAS, Kirsten rat sarcoma virus; MDT, multidisciplinary team; N/A, not applicable; NGS, next-generation sequencing; NSCLC, non-small cell lung cancer; PD-L1, programmed death-ligand 1; SMARCA4, SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4; STK11, serine/threonine kinase 11; VEGF, vascular endothelial growth factor.
Understanding of HER2 status across solid tumors and its predictive and prognostic role is evolving44,45
HER2 (ERBB2) is a known proto-oncogene first described in breast cancer.46 HER2 overexpression is a negative prognostic factor in multiple tumor types, including gastrointestinal (GI), endometrial, and ovarian.44
HER2 protein overexpression (score IHC 3+) may occur regardless of HER2 (ERBB2) amplification or HER2 (ERBB2) mutational status.47,48 Prevalence varies among tumor types but is associated with aggressive disease, poor outcomes, and increased recurrence risk.44,49,50
Prevalence of HER2 alterations across solid tumors
The prevalence of HER2 alterations varies in type of alteration and between tumor types.51
HER2 IHC 3+ (%) | HER2 IHC 2+ (%) | HER2 (ERBB2) amplificationa (%) | HER2 (ERBB2) mutationb (%) | |
---|---|---|---|---|
Breast52–59 | 10–20 | 19–49 | 13–23 | 2–4 |
NSCLC52,53,60–66 | 1–5 | 1–19 | ~1 | 2–4 |
Gastric/EGJ52,53,67–71 | 3–14 | 5–14 | 9–20 | 1–9 |
Biliary tract52,53,72–78 | 5–10 | 12–24 | 5–7 | 6–10 |
Pancreas53,56,59,77,79–81 | 1–7 | 6–9 | 2–24 | 1–3 |
Colorectum53,59,77,82–89 | 1–4 | 2–9 | 3–6 | 3–6 |
Bladder52,53,56,59,77,90–93 | 4–13 | 5–52 | 2–8 | 6–13 |
Endometrium53,56,59,77,94–97 | 3–28 | 14–39 | 4–8 | 2–3 |
Ovary52,53,56,59,77,98–101 | 1–5 | 0–24 | 1–2 | 1–3 |
Cervix52,53,56,59,102,103 | 4–11 | 14–18 | 3–7 | 2–5 |
aERBB2 (HER2) amplification is measured using ISH techniques, including FISH, CISH and SISH, or by NGS.45, 64b ERBB2 (HER2) mutations are detected through sequencing techniques, such as NGS or Sanger sequencing.63,83
HER2-positivity can enhance the metastatic potential of tumor cells104; a HER2-positive status may be a predictive factor, and HER2 testing is recommended for multiple metastatic solid tumors.105–116
Find out more about HER2 (ERBB2) mutations in mNSCLC.
Review testing guidelines
Follow testing recommendations in solid tumors with HER2 overexpression to inform treatment options
Test for HER2 by IHCÂ at metastatic diagnosis, to ensure that patients with a solid tumor receive personalized care.117,118
Summary of HER2 testing recommendations in guideline
Oncology specialty | Tumor type | Scoring criteria published | HER2 testing recommendations in the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) | HER2 tests included in tumor-specific CAP templates for reporting of biomarker testing results123–128 | ||
---|---|---|---|---|---|---|
HER2 overexpression | HER2 (ERBB2) amplification | HER2 (ERBB2) mutation | ||||
Breast | Breast | ASCO-CAP Guidelines118 | IHC, ISH108 | IHC | ISH | |
Gastrointestinal | Biliary tract | No universal guideline119 | IHC, FISH, NGS109 | General biomarker template availablec | ||
Gastric/EGJ | ASCO-ASCP-CAP Guidelines120 | IHC, ISH, NGS110,111 | IHC | ISH or Genomic test | Genomic test | |
Pancreatic | No universal guideline119 | IHC, NGS (amplification)112 | General biomarker template availablec | |||
Colorectal | HERACLES trial scoring criteria121 | IHC, FISH, NGS105,106 | IHC | ISH or Genomic test | Genomic test | |
Thoracic | NSCLC | No universal guideline119 | Sanger sequencing, targeted PCR and NGSÂ (mutation)113 | IHC | Genomic test | Genomic test |
Genitourinary | Bladder | No universal guideline119 | IHC114 | General biomarker template availablec | ||
Gynecologic | Endometrial | Fader trial scoring criteria122 | IHC, FISH107 | IHC | ISH | |
Ovarian | No universal guideline119 | IHC115 | General biomarker template availablec | |||
Cervical | No universal guideline119 | IHC, FISH116 | General biomarker template availablec |
aAssay-scoring system combinations should be validated according to the intended clinical use of the test, and the validation cohort should consist of at least 20 positive and 20 negative tissues.129Â bScoring criteria at discretion of laboratory director.129Â cWhere specific templates are not available, the CAP template for Reporting Results of Quantitive IHC Biomarker Testing of Specimens from Patients With Carcinoma may be used.128
ASCO-ASCP-CAP; American Society of Clinical Oncology-American Society for Clinical Pathology-College of American Pathologists; BRAF, v-raf murine sarcoma viral oncogene homolog B1; CCA, cholangiocarcinoma; CISH, chromogenic in situ hybridization; ERBB2, erb-B2 receptor tyrosine kinase 2; FISH, fluorescence in situ hybridization; GI, gastrointestinal; HER2, human epidermal growth factor receptor 2; IHC, immunohistochemistry; ISH, in situ hybridization; NCCN, National Comprehensive Cancer Network; NGS, next-generation sequencing; mNSCLC, metastatic non-small cell lung cancer; RAS, rat sarcoma; SISH, silver-enhanced in situ hybridization.
Most patients with breast cancer classified as IHC 0 have detectable levels of HER2 expression which could transform how we diagnose breast cancer.130–133
Evaluating HER2 IHC Expression Beyond Breast Cancer
ASCO-CAP and ASCO-ASCP-CAP have issued distinct guidelines for HER2 testing/scoring for breast and gastric cancers, defining distinct criteria for HER2-positivity by IHC (score IHC 3+) in the different tumor types. While circumferential membranous staining is required for an IHC score of 3+ for breast tumors, basolateral (“U”-shaped) membrane staining can be observed in IHC 3+ gastric tumors.119,121.Applying breast scoring criteria to gastric tumor specimens may lead to false-negative classification.119,121
HER2 expression extends beyond binary classification of positive or negative and may reveal biological differences130–136
HER2-negative is the most common breast cancer subtype, accounting for approximately 85% of all breast cancers.134 Despite being classified as HER2-negative, many of these tumors still carry some level of HER2 expression.135 It is estimated that approximately 60–65% of HER2-negative breast cancers are HER2-low.130,136
ASCO-ASCP-CAP, American Society of Clinical Oncology-American Society for Clinical Pathology-College of American Pathologists; HER2, human epidermal growth factor 2; IHC, immunohistochemistry.
Access educational resources, clinical cases and tools to evaluate your HER2 IHC scoring consistency in solid tumors.
Visit HER2Know.com
HER2, human epidermal growth factor 2; IHC, immunohistochemistry.

Considerations for Your Practice
Adopt cutting-edge improvements to help refine your MDT’s approach to biomarker and genetic testing and inform treatment decisions.
- Identify the right lab system and set up for your MDT
- Instigate laboratory training on digital pathology to improve workflow
- Aim for timelier diagnosis with computational pathology and deep learning algorithms
- Review screening protocols with the aim of detecting more cancers at treatable stagesÂ
- Monitor methods for ensuring patients are not lost to follow up post-treatment
- Align with guidelines on the latest biomarker testing recommendations
Explore solutions to common testing challenges encountered in your workflow
Hear from experts on best practice in biomarker testing
MDT, multidisciplinary team.

Subscribe to Emails
Subscribe and stay up to date with the latest updates in precision medicine.
Subscribe
Connect With Us
Contact our Precison Medicine Team for the latest developments to optimize your MDT's biomarker testing.
Connect with usMDT, multidisciplinary team.
References
- Bera K, et al. Nat Rev Clin Oncol. 2019;16(11):703–715.
- Chen RJ, et al. Cancer Cell. 2022;40(8):865–878.e6.
- Sha L, et al. J Pathol Inform. 2019;10:24.
- AstraZeneca. TrailblAZers in Oncology: Transforming patient outcomes through computational pathology. Accessed May 14, 2024. https://​www​.astrazeneca​.com/​media-centre/​articles/​2024​/oncology-trailblazers-digital-patholvogy.html
- Treanor D, et al. The Leeds Guide to Digital Pathology. Published online July 2018. Accessed June 20, 2024. https://​www​.leicabiosystems​.com​/sites​/default​/files​/media_document-file​/2022-01​/Brochure​-​Leeds​Guide​to​Digital​Pathology​%2818778​RevA%29.pdf
- Cui M, et al. Lab Invest. 2021;101(4):412–422.
- Cruz-Roa A, et al. Sci Rep. 2017;7:46450.
- Abels E, et al. J Pathol. 2019;249(3):286–294.
- Arvaniti E, et al. Sci Rep. 2018;8(1):12054.
- Saltz J, et al. Cell Rep. 2018;23(1):181–193.e7.
- U.S. Food and Drug Administration. FDA Authorizes Software that Can Help Identify Prostate Cancer. FDA.gov. Released September 21, 2021. Accessed May 16, 2023. https://www.fda.gov/news-events/press-announcements/fda-authorizes-software-can-help-identify-prostate-cancer
- Palm C, et al. Diagnostics (Basel). 2023;13(1):168.
- Wang S, et al. Cancers (Basel). 2019;11(11):1673.
- Campanella G, et al. Computer Vision and Pattern Recognition. 2022; arXiv:2206.10573 [cs.CV].
- Jeschke J, et al. Curr Opin Oncol. 2023;35(2):114.
- National Cancer Institute. Questions and Answers About MCD Tests. Accessed February 28, 2024. https://​prevention​.cancer​.gov​/major-programs​/multi-cancer-detection-mcd-research​/questions-and-answers-about-mcd-tests
- Etzioni R, Gulati R, and Weiss NS. J Natl Cancer Inst. 2022;114(3):349–352.
- Hubbell E, et al. Cancer Epidemiol Biomarkers Prev. 2021;30(3):460–468.
- National Cancer Institute. Dictionary of Cancer Terms – Minimal Residual Disease. Accessed February 28, 2024. https://​www​.cancer​.gov​/publications​/dictionaries​/cancer-terms​/def​/minimal-residual-disease
- Honoré N, et al. Cancers. 2021;13:5364.
- Peng Y, et al. Front Oncol. 2021;11:763790.
- Schuurhuis GJ, et al. Blood. 2018;131(12):1275–1291.
- Kumar S, et al. Lancet Oncol. 2016;17:e328–e346.
- Peng M, et al. Front Oncol. 2020;10:561598.
- Wu CY, et al. Diagnostics. 2020;10(3):144.
- Parsons HA, et al. Clin Cancer Res. 2020;26(11):2556–2564.
- McDonald BR, et al. Sci Transl Med. 2019;11(504):eaax7392.
- Obermayr E, et al. Oncotarget. 2017;8(63):106415–106428.
- Kuske A, et al. Sci Rep. 2016;6:39736.
- Novak D and Utikal J. Cancers (Basel). 2021;13(4):708.
- Sholl LM. Mod Pathol. 2022;35(suppl 1):66–74.
- Schoenfeld AJ, et al. Clin Cancer Res. 2020;26(21):5701–5708.
- Shang X, et al. Lung Cancer. 2021;154:105–112.
- Scalera S, et al. J Thorac Oncol. 2021;16(12):2065–2077.
- West HJ, et al. J Immunother Cancer. 2022;10(2):e003027.
- Skoulidis F, et al. Cancer Discov. 2018;8(7):822–835.
- Liu L, et al. Mol Oncol. 2021;15(2):462–472.
- Papillon-Cavanagh S, et al. ESMO Open. 2020;5(2):e000706.
- Liu R, et al. Nat Med. 2022;28(8):1656–1661.
- Cancer Genome Atlas Research Network. Nature. 2014;511(7511):543–550.
- Garcia-Robledo JE, et al. Ther Adv Respir Dis. 2022;16:1–35.
- Nadal E, et al. J Thorac Oncol. 2014;9(10):1513–1522.
- Alessi JV, et al. J Thorac Oncol. 2021;16(7):1176–1187.
- Iqbal N and Iqbal N. Mol Biol Int. 2014;2014:852748.
- Hanna WM, et al. Mod Pathol. 2014;27:4–18.
- Hechtman JF and Ross DS. Cytopathology 2019;127(7):428–431.
- Connell CM and Doherty GJ. ESMO Open. 2017;2:e000279.
- English DP, et al. Mol Diagn Ther. 2013;17(2):85–99.
- Reese DM and Slamon DJ. Stem Cells. 1997;15(1):1–8.
- Arteaga CL and Engelman JA. Cancer Cell. 2014;25(3):282–303.
- Cocco E, et al. Pharmacol Ther. 2019;199:188–196.
- Yan M, et al. Cancer Metastasis Rev. 2015;34(1):157–164.
- Uzunparmak B, et al. Ann Oncol. 2023;34(11):1035–1046.
- Farshid G, et al. Mod Pathol. 2020;33(9):1783–1790.
- Rakha EA, et al. Br J Cancer. 2021;124(11):1836–1842.
- Chmielecki J, et al. Oncologist. 2015;20(1):7–12.
- Owens MA. Clin Breast Cancer. 2004;5(1):63–69.
- Bose R, et al. Cancer Discov. 2013;3(2):224–237.
- Subramanian J, et al. Oncologist. 2019;24(12):e1303–e1314.
- Heinmöller P, et al. Clin Cancer Res. 2003;9(14):52385243.
- Zinner RG, et al. Lung Cancer. 2004;44(1):99–110.
- Takenaka M, et al. Anticancer Res. 2011;31(12):4631–4636.
- Zhuo X, et al. J Cancer Res Clin Oncol. 2023;149(5):2029–2039.
- Volckmar A-L, et al. Int J Cancer. 2019;145(3):649–661.
- Nagasaka M, et al. Clin Lung Cancer. 2022;23(1):52–59.
- Zhao J, et al. JCO Precis Oncol. 2020;4:411–425.
- Van Cutsem E, et al. Gastric Cancer. 2015;18(3):476–484.
- Subasinghe D, et al. J Int Med Res. 2023;51(2):1–16.
- Gao X, et al. Int J Surg. 2023;109(5):1330–1341.
- Im SA, et al. Diagn Mol Pathol. 2011;20(2):94–100.
- Cappellesso R, et al. Hum Pathol. 2015;46(5):665–672.
- Jeong H, et al. Cancers(Basel). 2021;13(2):161.
- Vivaldi C, et al. Oncologist. 2020;25(10):886–893.
- Roa I, et al. Gastrointest Cancer Res. 2014;7(2):42–48.
- Harder J, et al. World J Gastroenterol. 2009;15(36):4511–4517.
- Nakazawa K, et al. J Pathol. 2005;206(3):356–365.
- EEI Dumbrava, et al. JCO Precis Oncol. 2019;3:1–12.
- Li M, et al. Nat Genet. 2014;46(8):872–876.
- Chou A, et al. Genome Med. 2013;5(8):78.
- Han SH, et al. Diagnostics (Basel). 2021;11(4):653.
- Li X, et al. World J Surg Oncol. 2016;14:38.
- Cecchi F, et al. J Clin Oncol. 2023;41(4_suppl):194–194.
- Valtorta E, et al. Mod Pathol. 2015;28(11):1481–1491.
- Ingold-Heppner B, et al. Br J Cancer. 2014;111(10):1977–1984.
- Wang XY, et al. World J Gastrointest Oncol. 2019;11(4):335–347.
- Henry JT and Johnson B. Chin Clin Oncol. 2019;8(5):49.
- El-Deiry WS, et al. Cancer Biol Ther. 2015;16(12):1726–1737.
- Seo AN, et al. PLoS One. 2014;9(5):e98528.
- Raghav KPS, et al. Clin Cancer Res. 2024;30(2):260–262.
- Moustakas G, et al. J Int Med Res. 2020;48(1):1–12.
- Moktefi A, et al. Mod Pathol. 2018;31(8):1270–1281.
- Fleischmann A, et al. Eur Urol. 2011;60(2):350–357.
- Gårdmark T, et al. BJU Int. 2005;95(7):982–986.
- Vermij L, et al. Histopathology. 2021;79(4):533–543.
- Halle MK, et al. Br J Cancer. 2018;118(3):378–387.
- Buza N, et al. Mod Pathol. 2013;26(12):1605–1612.
- Semiz HS, et al. Turk Patoloji Derg. 2023;39(1):55–63.
- Tuefferd M, et al. PLoS One. 2007;2(11):e1138.
- Ersoy E, et al. Int J Gynecol Pathol. 2022;41(4):313–319.
- Chung YW, et al. J Gynecol Oncol. 2019;30(5):e75.
- Pils D, et al. Br J Cancer. 2007;96(3):485–449.
- Shi H, et al. J Pathol Clin Res. 2021;7(1):86–95.
- Panek G and Ligaj M. Gin Oncol. 2007;5(4):218–235.
- Cheng X. Genes. 2024;15(7):903.
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Colon Cancer V.5.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Rectal Cancer V.4.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Uterine Neoplasms. V.3.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Breast Cancer V.6.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed November 11, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Biliary Tract Cancers V.5.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed Nov 27, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Gastric Cancer V.4.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Esophageal and Esophagogastric Junction Cancers V.4.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Pancreatic Adenocarcinoma V.3.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Non-Small Cell Lung Cancer V.11.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 23, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Bladder Cancer V.5.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed November 07, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Ovarian Cancer Including Fallopian Tube Cancer and Primary Peritoneal Cancer V.3.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Cervical Cancer V.4.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed October 01, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org
- Vranic S and Gatalica Z. Acta Med Acad. 2021;50(1):47–57.
- Wolff AC, et al. Arch Pathol Lab Med. 2023;147(9):993–1000.
- Zhang H, et al. Histopathology. 2024. doi: 10.1111/his.15170.
- Bartley AN, et al. J Clin Oncol. 2017;35(4):446–464.
- Sartore-Bianchi A, et al. Lancet Oncol. 2016;17(6):738–746.
- Fader AN, et al. J Clin Oncol. 2018;36(20):2044–2051.
- CAP template for Reporting Results of HER2 (ERBB2) Biomarker Testing of Specimens from Patients with Adenocarcinoma of the Stomach or Gastroesophageal Junction. Version 1.0.0.1. Available at: https://documents.cap.org/protocols/cp-gastric-HER2biomarker17-1001.pdf. Accessed: February 2024.
- CAP Template for Reporting Results of Biomarker Testing of Specimens From Patients With Carcinoma of the Colon and Rectum. Version 1.3.0.0. Available at: https://documents.cap.org/protocols/ColoRectal.Bmk_1.3.0.0.REL_CAPCP.pdf. Accessed June 10, 2024.
- CAP Template for Reporting Results of Biomarker Testing of Specimens From Patients With No-Small Cell Carcinoma of the Lung. Version 2.0.1.1. Available at: https://documents.cap.org/protocols/Lung.Bmk_2.0.1.1.REL_CAPCP.pdf. Accessed June 10, 2024.
- CAP Template for Reporting Results of Biomarker Testing of Specimens From Patients With Carcinoma Gynecologic Origin. Version 1.1.0.0. Available at: https://documents.cap.org/documents/Gynecologic.Bmk_1.1.0.0.REL_CAPCP.pdf. Accessed June 10, 2024.
- CAP Template for Reporting Results of Biomarker Testing of Specimens from Patients with Carcinoma of the Breast. Version 1.5.0.1. Available at: https://documents.cap.org/documents/Breast.Bmk_1.5.0.1.REL_CAPCP.pdf. Accessed June 10, 2024.
- CAP Template for Reporting Results of Quantitative IHC Biomarker Testing of Specimens From Patients with Carcinoma. Version 1.1.0.1. Available at: https://documents.cap.org/protocols/IHC.Bmk_1.1.0.1.REL_CAPCP.pdf. Accessed June 10, 2024.
- Goldsmith JD, et al. Arch Pathol Lab Med. 2024;148:e111–e153.
- Chen Z, et al. Breast Cancer Res Treat. 2023 Nov;202(2):313–323.
- Boyraz B, Ly A. Hum Pathol. 2022;127:50–55.
- Ardor GD, et al. Breast J. 2023;2023:9725647.
- Venetis K, et al. Front Mol Biosci. 2022;9:834651.
- Tarantino P, et al. J Clin Oncol. 2020;38(17):1951–1962.
- Sajjadi E, et al. Cancer Drug Resist. 2022;5(4):882–888.
- Denkert C, et al. Lancet Oncol. 2021;22(8):1151–1161.
NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way.