CC BY 4.0 · Indian J Med Paediatr Oncol 2025; 46(03): 233-252
DOI: 10.1055/s-0044-1788727
ISMPO Guidelines

Genetic Counselling, Testing, and Management of Hereditary Breast and Ovarian Cancer Syndrome in India: Updated Expert Consensus Recommendations from Indian Society of Medical and Pediatric Oncology

1   Department of Medical Oncology, Sri Ram Cancer Center, Mahatma Gandhi Medical College Hospital, Jaipur, Rajasthan, India
,
2   Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, India
,
3   Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
,
Pradyna Kotwal
4   Department of Recombinant DNA Facility, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
,
5   Department of Medical Oncology, Cancer Institute (WIA - Women's Indian Association), Chennai, Tamil Nadu, India
,
6   Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
,
Thangarajan Rajkumar
7   Department of Molecular Oncology, MedGenome, Bangalore, Karnataka, India
,
8   Department of Cancer Genetics, Tata Memorial Hospital, Mumbai, Maharashtra, India
,
9   Department of Medical Oncology, Healthcare Global Enterprises Ltd. Hospital, Bengaluru, Karnataka, India
,
Ajay Bapna
10   Department of Medical Oncology, Bhagwan Mahaveer Cancer Hospital & Research Centre, Jaipur, Rajasthan, India
,
B. K. Smruti
11   Department of Medical Oncology, Lilavati Hospital and Bombay Hospital Institute of Medical Sciences, Mumbai, Maharashtra, India
,
Rupinder Sekhon
12   Department of Gynae Oncology, Artemis Cancer Center, Gurugram, Haryana, India
,
Maheboob Basade
13   Department of Medical Oncology, Saifee Hospital, Mumbai, Maharashtra, India
,
Sabhayata Gupta
14   Department of Gynae Oncology, Medanta the Medicity, Gurugram, Haryana, India
,
Amita Maheshwari
15   Department of Gynae Oncology, Tata Memorial Centre and Homi Bhabha National Institute, Maharashtra, Mumbai, India
,
S. P. Somashekhar
16   Department of Surgical Oncology, Aster International Institute of Oncology, Bengaluru, Karnataka, India
,
17   Department of Surgical Oncology, Max Healthcare, New Delhi, Delhi, India
,
Arvind Krishnamurthy
18   Department of Surgical Oncology, Cancer Institute, Chennai, Tamil Nadu, India
,
Anushree Punia
19   Department of Radiation Oncology, Shri Ram Cancer Centre, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, India
,
Moushumi Suryavanshi
20   Department of Molecular Biology, Amrita Institute of Medical Sciences and Research Centre, Faridabad, Utter Pradesh, India
,
Rekha Gupta
21   Department of Medical Genetics, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, India
,
Amit Verma
22   Department of Molecular Oncology, Dr. AV Cancer Institute, Gurgaon, Haryana, India
,
Padmaj Kulkarni
23   Department of Medical Oncology, Deenanath Mangeshkar Hospital, Pune, Maharashtra, India
,
Purvish Parikh
24   Department of Clinical Hematology, Mahatma Gandhi University of Medical Sciences and Technology, Jaipur, Rajasthan, India
› Institutsangaben
 

Abstract

Introduction Hereditary breast and ovarian cancer (HBOC) is driven by mutations in BRCA1/2 and related genes. Their understanding is vital to appropriate management of such patients and at-risk families, including counselling and genetic testing. Several important recent advances have made it necessary to revise the previous recommendations we made for India in 2020.

Materials and Methods This consensus document was developed with the authors as key experts in the field. Published evidence, real-world data, and expert interpretation were used by a modified Delphi method to finalize these recommendations.

Results Detailed description and process for identifying patients at risk, doing their counselling, selecting the right molecular test, interpreting the results, and determining the optimal mode of action to attenuate risk of HOBC or its recurrence have been provided in a clear and lucid manner. Differences between germline and somatic mutations are described. Information from publicly available databases was used to fine-tune the guidelines—as more information had becomes available since the time of writing the first guidelines. Risk of various cancer types and corresponding risk reduction strategies have been explained.

Conclusion Community oncologists in India, SAARC region, and other low- and middle-income countries should use these guidelines in their clinical practice to optimize genetic counselling, molecular testing, and management of patients with HBOC.


#

Introduction

In India, breast cancer (BC) is still the commonest cancer in females as well as the leading cause of cancer-related death in women.[1] While the younger median age of onset reflects the population demographics (52% of Indians are below the age of 30 years), BC in Indian women has a high incidence-to-mortality ratio compared with the West.[2] It is therefore important to ascertain risk factors, high-risk biomarkers, and driver mutations to optimize management.

As the name suggests, hereditary breast and ovarian cancer (HBOC) syndrome leads to increased risk of early-onset BC and ovarian cancer (OC) in multiple family members. It usually follows an autosomal-dominant inheritance pattern.[3] [4] HBOC syndrome results in higher lifetime risk of cancer in women—being 50 to 85% for BC, and 15 to 30% for OC.[5] [6] Most common mutations responsible for HBOC include BRCA1 and BRCA2.[7] Race and ethnicity have strong bearing in HBOC. For instance, specific mutations identified in a population sharing common ancestry (Founder mutations) first reported from the Caucasians were from Ashkenazi Jews, French Canadians, and Icelanders.[8] Similarly novel founder mutations in BRCA1, BRCA2, and MLH1 genes have been identified in India from several Maharashtrain, Gujarati, Jain, Bohri, Punjabi, Bengali, and Nepali communities[9] [10] [personal communication Rajiv Sarin, 27 October 2023].

With the availability of drugs that have been proven to be useful in women with BRCA1/2 mutations (e.g., poly (ADP-ribose) polymerase [PARP] inhibitors for both germline and somatic mutations; platinum-based chemotherapy in germline), their genetic testing is vital for optimal treatment decision making.[11] Identification of such mutations (and families at risk of HBOC) allows us to quantify their risk of future metachronous cancers as well as discuss pros and cons of appropriate surgical and/or nonsurgical prophylactic measures.[12] We can then enable families at risk to become informed previvors (a person who takes action to reduce or eliminate a genetic cancer before the cancer develops or is detected in his or her body).[13] We now know that “BRCA-ness” is also a result of mutations in several non-BRCA genes (e.g., PALB2, CHEK2, ATM1, RAD51C, and RAD51D) and similar preventive measures can also potentially benefit them also.[14]

The prevalence, nature, and frequency of germline mutations vary significantly—geographically and ethnically.[8] [9] Their clinical significance and penetrance is also highly variable. BrCa Exchange collates data from across the world, including India, and makes them publicly available for real-time risk assessment for individual patient–related genetic mutations. In general, pathogenic genetic mutation occurs in about 10 to 15% of all patients with BC, and BRCA1 and BRCA2 account for almost half of the pathogenic/likely pathogenic mutations.[9] [10] [15] [16]

While innumerable international guidelines have been published since 2010 for overall management of HBOC, almost all of them are based on the Caucasian population.[17] Our 2020 consensus document was the first to incorporate unique characteristics and needs of the Indian patients.[19]

In the intervening period, a lot of new data have been generated and our understanding of HBOC refined. It was therefore necessary to update our previous recommendations, making it more robust.


#

Materials and Methods

Our multidisciplinary expert group began the revision process by re-evaluating evidence from phase III randomized controlled studies, other prospective and retrospective studies, BrCa gene mutation publications, and real-world clinical experience. Google Scholar and PubMed databases were searched with the key words: “hereditary breast and ovarian cancer”; “HBOC”; “BRCA1/2 mutations”; “germline BRCA mutations”; “somatic BRCA mutations”; “brcaness”; “non-BRCA mutations”; and “genetic testing.”

The first meeting involved extensive discussions moderated by the chairperson of the committee followed by preliminary voting using the modified Delphi process.[20] Thereafter, the Expert Committee continued to discuss via e-mail and WhatsApp to fine-tune the recommendation statements. All authors participated in the preparation of the draft, multiple rounds of voting, and the approval of the final manuscript.


#

Recommendations (Results)

1. Genetic Counseling in India: Importance and Awareness

While our understanding has improved considerably, genetic testing in HBOC in India lags behind for several reasons, most important ones being infrastructure and finances.[21] Lack of sufficient number of trained genetic counsellors also increases the burden of the treating physician and oncologist. Where feasible, individual or family members are helped appropriately using the following principles:

  • Allow them to understand the medical facts in simple terms: diagnosis, probable future course, cancer risk, and management options.

  • Understand role of heredity in the risk of cancer, its risk of recurrence in patients, and first-degree relatives who are carrying the mutation(s).

  • Understand how to minimize the risk of recurrence.

  • Select the course of action most suitable to their individual preference and family goals.

  • Understand the advantages of positive lifestyle changes.[22] [23] [24] [25]

Pretest genetic counselling is generally recommended globally. This is also endorsed by international oncology working groups.[4] [26] [27] [28] Such counselling should be done by health care professionals who are adequately trained with respect to genetic and clinical aspects of HBOC, BRCA1 and BRCA2.[29] [30] In India, the burden often falls on the respective oncologists.[21] [31] In view of the shortage of trained and qualified staff across India, use of tele-genetic counselling is recommended. This should be carried out in compliance with the Telemedicine Practice Guidelines as published by Government of India.

Components of HBOC Genetic Counselling

  • (A) Pretest counselling: the patient/family members should be made aware of the following key factors:

    • Medical history and available pedigree evaluation of up to three generations ([Fig. 1]).

    • Objective risk estimation using mathematical risk assessment models where available and qualitative criteria as an alternate (e.g., National Comprehensive Cancer Network [NCCN] tools).

    • Genetic testing methodology recommendations.

    • Interpreting test reports—possible test outcomes, e.g., pathogenic variant detected, variant of uncertain significance (VUS).

    • Risks, benefits, and psychosocial implications.

    • Implications of test results to other family members.

    • Discussion of need to maintain confidentiality to prevent genetic discrimination.[22] [24] [25]

    Once the pretest genetic counselling is documented, if the patient/subject allows to be subject to biological sample collection, consent is automatically implied.

    Assessment of family history: while real-world limitations are well known in countries like India, attempts should be made to collect family history of three generations to prepare pedigree analysis. This should ideally include first- and second-degree relatives on both the maternal and paternal sides of the family. Details recommended are age, gender, pregnancies (if applicable), age at onset of cancer (if applicable), age at death (if applicable), cause of death (for deceased relatives), ethnic background of grandparents (maternal and paternal, if necessary), and history of consanguinity. Using this information to generate a pedigree chart gives a good visual understanding for all stake holders.[32] [33] [34] [35]

  • (B) Post-test counselling : the key objective of this session is risk communication. Once the molecular testing report becomes available and the health care team has calculated the risk of cancers/recurrence, the same is communicated to the patient/family member as appropriate. Appropriate attention should also be given to emotional counselling. Pretest genetic counselling improves the family's journey through this process. Group discussions and use of information booklets, where available, are encouraged. This can minimize anxiety of the patients and their family members.[33] [34] [35] [36] It is desired that all questions and doubts are answered to the satisfaction of the patient/family member during this session.[37]

Zoom Image
Fig. 1 Representative pedigree chart of HBOC family. HBOC, hereditary breast ovarian cancer.

#
#

2. Germline BRCA Testing

Assessment of risk and identifying patients: germline mutations in BRCA1/2 genes are regarded as high penetrance—a cancer relative risk of greater than 5—and have been characterized in several populations globally. Mutations in other non-BRCA genes, such as PALB2, TP53, PTEN, CDH1, STK11, CHEK2, RAD51C, RAD51D, and ATM, are also known to confer risk of BC and/or OC, albeit with lower frequency and penetrance[26] [31] [35] ([Tables 1] and [2]).

Table 1

Genes associated with HBOC and their penetrance risk

Gene/locus

Syndrome

Risk

Mutation/minor allele frequency

High penetrance

 BRCA1 (17q21)

HBOC

60–85 × for BC

1/400

15–40 × for OC

 BRCA2 (13a12.3)

HBOC

60–85 × BC

1/400

13–23 × OC

 TP53 (17p13.1)

Li–Fraumeni

50–89 × by age 50 BC

<1/10,000

90 × in Li–Fraumeni survivors

 PTEN (10q23.3)

Cowden

25–50 × BC

<1/10,000

 CDH1 (16q22.1)

Familial diffuse gastric cancer

RR: 6.6

<1/10,000

 STK11/LKB1 (19p13.3)

Peutz–Jeghers

30–50 × by age 70

<1/10,000

 PALB2 (16p12)

BC, prostate Ca, pancreatic Ca

30% BC risk by age 70

<1/1,000

Moderate penetrance

 CHEK2 (22q12.1)

Li–Fraumeni 2

OR: 2.6

1/100–1/200 in some populations

 BRIP1 (17q22)

BC

RR: 2.0

<1/1,000

 ATM (11q22.3)

Ataxia telangiectasia

RR: 2.37

1/33–1/333

Low penetrance

 FGFR2 (10q26)

BC

OR: 1.26

0.38

 TOX3 (16q12.1)

BC

OR: 1.14

0.46

 LSP1 (11p15.5)

BC

OR: 1.06

0.3

 TGFB1 (19q13.1)

BC

OR: 1.07

0.68

 MAP3K1 (5q11.2)

BC

OR: 1.13

0.28

Abbreviations: BC, breast cancer; HBOC, hereditary breast ovarian cancer; OC, ovarian cancer; OR, odds ratio; RR, relative risk.


Table 2

Cancer risks associated with BRCA1 and BRCA2

Mutation

Lifetime risk of BC, %

Lifetime risk of OC, %

 BRCA 1

50–85

35–46

 BRCA 2

50–85

13–23

Cancer type

Risk in carriers to age 70 years

Lifetime risk in general population, %

 Breast

BRCA1: 55–70

BRCA2: 45–70

 Contralateral breast

Up to 63 at 25 years post-diagnosis, but highly age-dependent

7 at 25 years post-diagnosis

 Ovarian

BRCA1: ∼40

∼1

BRCA2: ∼15

 Colon

Unclear

∼5

 Prostate

Elevated; absolute risk not well defined

White: ∼14

African American: ∼19

 Male breast

BRCA1: 1

0.1

BRCA2: 8

 Pancreatic

BRCA1: unclear

1.5

BRCA2: 5

 Other sites

To be determined

Varied

Abbreviations: BC, breast cancer; OC, ovarian cancer.


Source: Adapted from Malhotra et al[19].


The lifetime risk of breast and ovarian malignancies is variable, with pathogenic mutations in BRCA1 (BC: 46–87%; OC: 39–63%) and BRCA2 (BC: 38–84%; OC: 17–27%). Other cancers associated with germline BRCA1/2 mutations include male BC (1–9%), prostate cancer (9–20%), pancreatic cancer (1–7%), and melanoma.[38] The largest analysis of 1,010 high-risk families across India revealed BRCA mutations in 85% and non-BRCA mutations in 15% of families.[39] Additional analysis based on age and family history showed a high prevalence of germline variants (75%) in younger patients age younger than 40 years with a first-degree family member affected with BC/OC.[40] [41] [42] A methodical review investigating the prevalence of germline variants in high-risk HBOC susceptibility genes in 1,028 patients of Indian descent with familial/early-onset/triple-negative BC (TNBC) or OC identified 18 BRCA1 and 16 BRCA2 variants that were not reported in the Breast Cancer Information Core or ClinVar databases.[39] The putative Ashkenazi founder mutation BRCA1 185delAG was detected in a low proportion of patients (4.2%), the majority of whom were from South India or who were Malaysians of Indian origin.[40] [41] [42] In the last 5 years, several studies have been published from Indian researchers investigating the frequency of P/LP mutations in the Indian population. [Table 3] provides a summary of the important Indian-specific studies published till date. Most of the studies have a strong selection bias in patient enrollment and hence have reported higher frequencies.[39] [43] [44] [45] [46] [47] [48] However, two studies have been performed in unselected patients with BC and OC[39] [43] In a predominantly North Indian population, Mittal et al have reported a frequency of 18.6% for P/LP variants, with their multigene next-generation sequencing (NGS) panel.[44] Using the NCCN 2019 criteria would have missed 11% of these mutations. This frequency is higher than those reported on unselected cases from Europe, the United States, and Africa. In their multicentric study, Gupta et al tested 239 unselected patients with OC for only BRCA1/2. They reported a prevalence of 21.4% P/LP mutations.[43] It is also important to keep in mind that NGS testing has the risk of leading to false positivity. In one study, out of all 41 samples analyzed for BRCA1 and BRCA2, 5 were found with 950_951 insA (Asn319fs) at Chr13:32906565 position and 1 sample with 1032_1033 insA (Asn346fs) at Chr13:32906647, both being frame-shift mutations in BRCA2 gene. The 950_951 insA (Asn319fs) mutation is reported as a pathogenic allele in NCBI dbSNP. On examination of IGV for all these samples, it was found that both mutations had “A” nucleotide insertion at 950 and 1032 positions in exon 10 of BRCA2 gene. However, Sanger sequencing did not confirm these insertions.[49] [50] [51] [52] [53]

Table 3

Pathogenic BRCA1/2 mutations identified in Indian patients

Study

Region

Tumor types

Testing method

Remarks

Valarmathi et al, 2003[118]

New Delhi, North India

Breast cancer

Direct sequencing

BRCA 1 (E1250X in exon 11; E1754X in exon 20)

Rajkumar et al, 2003[79]

Chennai, South India

Breast and ovarian cancer

Heteroduplex analysis/dHPLC

BRCA1 (Ex12 1386 delCTCTC Stop 1389, Ex13 CGA → TGA Arginine 1443 Stop), BRCA2 (Ex11O 1235delCTTAA stop 1237)

Saxena et al, 2006[119]

New Delhi, North India

Breast cancer

Heteroduplex analysis of PCR amplicons using exon-specific primers

BRCA1 (185delAG in exon 2; 4184del4; 3596del4 in exon 11), BRCA1 (4184del4 in exon 11)

Syamala et al, 2007[120]

Kerala, South India

Breast and ovarian cancer

Direct sequencing

BRCA2 (c.4642delAA, c.4926insGACC)

Thirthagiri et al, 2008[42]

Malaysia, Indian ethnicity

Breast cancer

dHPLC and DNA sequencing

BRCA1 (180 delA, 185 delAG, 5370 C.T), BRCA2 (9097 C.T)

Soumittra et al, 2009[121]

Chennai, South India

Breast and ovarian cancer

PCR-dHPLC

BRCA1 (c.4158_4162delCTCTC; p.Ser1369SerfsX2, c.4327C.T; p.R1443X, c.1148_1149delAT; p.Asn383Arg fsX6, c.4399C.T; p.Gln1467X, c.4705_4706insTGGAATC; p.Ile1567fsx5, c.5024_ 5025insT; p. Thr1675Thr fsX4, c.68_69delAG; p.Glu23Val fsX16, c.66_67delAG; p. Leu22Leu fsX18, c.5118_5120delAAT; p.del1707Ile); BRCA2 (c.6214_6218delCTTAA; p.Ser2072Ser fsX4, c.5130_5133delTGTA; p.Tyr1693X, c.2621_ 2627delAACTGTC; p. Ile873Ile fsX19)

Vaidyanathan et al, 2009[55]

South India

Breast and ovarian cancer

Heteroduplex analysis using CSGE and direct sequencing

BRCA1 (185delAG)

Kang et al, 2014[18]

Malaysia, Indian ethnicity

Breast cancer

PCR and Sanger sequencing

BRCA1 (185delAG)

Mittal et al, 2022[44]

North India

Breast cancer

NGS, multigene panel with reflex MLPA

236 unselected, consecutive patients, 18.64% P/LP, 34% non-BRCA genes, 1 AJ founder mutation

Pramanik et al, 2022[45]

North India

Ovarian cancer

NGS, multigene panel with reflex MLPA

72 patients, selectively referred for genetic testing. 44% P/LP, 85% BRCA, and 15% non-BRCA

Gupta et al, 2021[43]

Pan India

Ovarian cancer

NGS, BRCA1/2

239 unselected patients, 21.4% P/LP

Kadri et al, 2021[46]

Western India

Breast and ovarian cancer

NGS, multigene panel

144 patients, selectively referred, 28% P/LP, 12% non-BRCA.

Singh et al, 2018[39]

Pan India

Breast and ovarian cancer

NGS, multigene panel

1,010 selectively referred, 30.1% P/LP, BRCA1/2 85%

Chheda et al, 2020[47]

Western India

Breast and ovarian cancer

NGS, BRCA1/2

160 women, selectively referred, 31.9% P/LP

Mehta et al, 2018[48]

North India

Breast and ovarian cancer

NGS, BRCA1/2

206 women, selectively referred, 30.6% P/LP

Abbreviations: MLPA, multiplex ligation-dependent probe amplification; NGS, next generation sequencing.


Clinical practice has been focused to test patients who fulfil NCCN criteria for testing ([Box 1]); however, recent publications have emphasized that using NCCN guidelines misses many patients with both BRCA and non-BRCA mutations.[54] At present, it is therefore uncertain whether testing for hereditary mutations is warranted beyond the standard criteria. As a rule of thumb, it is recommended to assess women with a personal or family history of BC, OC, tubal, or peritoneal cancer or those who are part of a family having known BRCA1/2 gene mutations. Where appropriate, a familial risk assessment tool should guide whether further genetic counselling and/or genetic testing is warranted.[55]

Box 1

Guidelines for gBRCA risk assessment (adapted from NCCN Criteria v.3 2023)

1. Personal history of breast cancer with specific features:

 • ≤50 years

 • Any age:

  ◊ Treatment indications

   – To aid in systemic treatment decisions using PARP inhibitors for breast cancer in the metastatic setting

   – To aid in adjuvant treatment decisions with olaparib for high-risk, HER2-negative breast cancer

 ◊ Pathology/histology

   – Triple-negative breast cancer

   – Multiple primary breast cancers (synchronous or metachronous)

   – Lobular breast cancer with personal or family history of diffuse gastric cancer

 ◊ Male breast cancer

 ◊ Ancestry: Ashkenazi Jewish ancestry

 ◊ Family history

   – ≥1 close blood relative with ANY:

    ▪ breast cancer at age ≤50

    ▪ male breast cancer

    ▪ ovarian cancer

    ▪ pancreatic cancer

    ▪ prostate cancer

  – ≥3 total diagnoses of breast cancer in patient and/or close blood relatives

   – ≥2 close blood relatives with either breast or prostate cancer (any grade)

2. Personal history of epithelial ovarian cancer (including fallopian tube cancer or peritoneal cancer) at any age

3. General criteria:

 • Individuals with any blood relative with a known P/LP variant in a cancer susceptibility gene

 • Individuals meeting the criteria below but who tested negative with previous limited testing

 • A P/LP variant identified on tumor genomic testing that has clinical implications if also identified in the germline

 • To aid in systemic therapy and surgical decision-making

• Individual who meets Li–Fraumeni syndrome (LFS) testing criteria or Cowden syndrome/PTEN hamartoma tumor syndrome (PHTS) testing criteria or Lynch syndrome

. Family history of cancer only:

 • An affected individual (not meeting testing criteria listed above) or unaffected individual with a first- or second-degree blood relative meeting any of the criteria listed above (except for systemic therapy decision-making).

 • If the affected relative has pancreatic cancer or prostate cancer only, first-degree relatives should be offered testing unless indicated based on additional family history.

 • An affected or unaffected individual who otherwise does not meet the criteria above but has a probability >5% of a BRCA1/2 pathogenic variant based on prior probability models (e.g., Tyrer–Cuzick, BRCAPro, CanRisk)


#
#

Whom to Test First?

Any woman affected by early-onset BC and/or OC should be tested first. A suspected family member who is below the age of 18 years should have testing differed till they become adults. Once an index case (proband) has been identified, testing of first-degree adult relatives is the next step. If a pathogenic variant/mutation has already been identified, single-site gene testing of the specific gene mutation is sufficient. This is called cascade testing (screening of at-risk biologic relatives of the individual who harbors a pathogenic variant).[56] Its objective is to identify asymptomatic at-risk relatives, calculate their risk, and discuss steps to reduce future morbidity and mortality from cancer (risk-reducing strategies include enhancing surveillance diagnostics, medical therapeutics, and/or surgical interventions).

Testing Methods

First-generation automated Sanger sequencing has generally been replaced by the more efficient NGS technology. It is less expensive and comprehensive genomic profiling or multigene panel testing becomes rapid. Where cost is not a significant consideration, multigene panels are being used as a first-line test for any patient suspected to have an inherited cancer syndrome.[57]

This approach is robust for detecting single-nucleotide variants (SNVs) and small insertion/deletion (indels), with high accuracy. However, this is not the case for larger genomic rearrangements (insertions/deletions) and/or copy number variants. Multiplex ligation-dependent probe amplification (MLPA) or array comparative genomic hybridization (aCGH) would have to be used to detect the same.[58] Several in silico tools make the bioinformatics part less labor-intensive and reduce turnaround time.

Based on genetic testing and interpretation, patients can be divided into three risk groups—high risk, moderate risk, and low or unknown risk ([Table 1]).


#

Methods of Germline BRCA Detection

Sample for germline genetic testing could be blood, saliva, or cheek swab. As mentioned above, multigene panels using NGS enables high-throughput genetic testing with good accuracy. However such testing can be performed as follows:

  • Single site.

  • Founder/recurrent mutation associated with specific neoethnicity.

  • Multigene/targeted panel.

  • MPLA/aCGH for large genomic rearrangements/copy number variations.

  • Whole exome sequencing.

  • Whole genome sequencing.

While selecting a NGS workflow, the following criteria should be considered to suit the genetic testing[59] [60] [61] ([Box 2]):

Box 2

Points to be considered while selecting the right lab for genetic testing:

1. Certification (e.g., NABL)

2. Participation in external quality assurance program

3. Documentation of internal quality control SOPs

4. Design of appropriate multigene panel

5. Wet lab processes for enrichment of targets (hybrid capture or amplicon-based capture) without compromising on sensitivity and specificity of the detection of pathogenic variants

6. Bioinformatic expertise

7. Qualified and trained personnel to interpret results

8. Reasonable turnaround time (approximately 4 weeks)

  • Design of multigene panel based on clinical utility and demand.

  • Wet lab processes for enrichment of targets (hybrid capture or amplicons based capture) without compromising on sensitivity and specificity of the detection of pathogenic variants.

  • Bioinformatics expertise.

  • Qualified and trained personnel to interpret results.

  • Reasonable turnaround time (approximately 4 weeks).

Having a reasonable turnaround time (approximately 4 weeks) helps in reducing anxiety and testing fatigue amongst patients, families, and providers.

At least three studies from India have reported the use of multigene panel testing by NGS for germline mutations in HBOC patients.[39] [43] [44] [45] [46] [47] [48] [49] The majority of BRCA1/2 mutations identified are single base substitutions (missense or nonsense mutations); small insertions or deletions (result in prematurely truncated nonfunctional protein); and splice junction alterations (exon skipping or intronic inclusion, also resulting in a nonfunctional protein). In 5% of cases, there could be large genomic rearrangements which will be missed on conventional NGS testing. As mentioned above, MLPA technique or aCGH can be used in patients/families who are shown negative by NGS, but have a strong clinical suspicion of HBOC.[48] [49] It is important to remember that besides BRCA1 and BRCA2, genes that need to be tested in suspected cases of HBOC syndrome includes ATM, BRIP1, CHEK2, RAD50, RAD51D, RAD51C, PALB2, BAARD1, P53, STK11, CDH1, MSH2, MSH6, MLH1, EPCAM, PMS2, ATM, PTEN, FGFR2, TOX3, LSP1, and MAP3K1.


#

Interpretation of Sequencing Results

International working groups have provided guidelines for the interpretation of germline sequence variants. The DNA sequence alterations are categorized qualitatively based on several factors—including functional evidence, family history, allele frequency data, computational and in silico predictions ([Table 4]).

Table 4

Classification of mutation variants according to clinical significance (adapted from nomenclature of three international working groups)

International Agency for Research in Cancer

Clinical Molecular Genetics Society

American College of Medical Genetics

Class

Description

Class

Description

Category

Description

1

Definitely pathogenic

1

Certainly nonpathogenic

1

Previously reported and recognized cause of the disorder

2

Likely pathogenic

2

Unlikely to be pathogenic

2

Previously unreported and is of the type that is expected to cause the disorder

3

Uncertain

3

Likely to be pathogenic

3

Previously unreported and is of the type that may or may not be causative of the disorder

4

Likely not pathogenic

4

Certainly pathogenic

4

Previously unreported and is probably not causative of disease

5

Not pathogenic

5

Previously reported and is a recognized neutral variant

6

Previously not known or expect to be causative of disease, but is found to be associated with a clinical presentation

Source: Adapted from Malhotra et al[19].


Failure to detect a deleterious germline mutation in a proband could be due to one or more of the following[49] [62]:

  • Patient has a pathogenic variant in another gene not included in the multigene panel.

  • Tested gene has a sequence variant that cannot be easily detected by sequence analysis (e.g., large deletion; limitation of test methodology).

  • Sequence variant in a region such as an intron or regulatory region of a gene (area not covered by the test).

  • Involvement of genes not associated with known underlying phenotype.

Interpretation is facilitated by interrogating updated publicly available literature/databases (ClinVar, OMIM, BrCa Exchange, GWAS, HGMD, and SwissVar), correlation with population data, and in silico predictions of variant effect. Nonsynonymous variants' effects can be calculated using multiple algorithms such as PolyPhen-2, SIFT, Mutation Taster2, Mutation Assessor, and LRT. Only nonsynonymous and splice site variants found in the hereditary cancer gene panel should be used for clinical interpretation. Fortunately, germline BRCA testing is robust and the variants are well-curated.


#

Genetic Test Report

Molecular labs should transcribe their information into a report that describes the test results in a uniform manner, without using jargon, and facilitates explaining its significance to the proband and first-degree relatives. DNA change as a variant should be reported using the standard Human Genome Variation Society (HGVS) nomenclature, describing the mRNA reference sequence that was used, the nucleotide change in the cDNA as a c. and the consequent change in the amino acid and protein as a p.[63] [64]


#

Validation of Test Result

Molecular testing laboratories should follow the joint consensus from the Association for Molecular Pathology and College of American Pathologists. They should validate every detected SNV or indel in the coding region that results in deleterious mutations and documenting it in terms of positive percentage agreement and positive predictive value.[64] [65] Documenting concordance between results from a newly developed assay and a gold standard method such as Sanger sequencing is also advised. Participating in internal quality control as well as external quality assurance programs is desirable. If using the NGS for genetic testing, then validating NGS results by Sanger may not be necessary if all bioinformatic and quality parameters are controlled.[43] [44] [49] [66]


#
#

How to Manage Variants of Uncertain Significance

VUSs are genetic alterations whose clinical significant is not yet established. They are usually single nucleotide polymorphisms—may be in the promoter regions, exons, and introns. They could also be small in frame insertions and deletions or synonymous substitutions.[64] More than 20,000 unique variants have been identified in the BRCA genes.[65] These constitute less than 10% of all mutations in BRCA1/2 (the remaining 90% have already been classified either as pathogenic or benign). The clinical implication is that once further data become available, up to 30% of current VUSs might turn out to be pathogenic.[67] [68] In countries where data are still maturing, like India, up to half (30–50%) of mutations identified in BRCA1/2 genes could be VUS.[9] Data sharing initiatives like BrCa Exchange, BRCA Challenge, and Evidence-based Network for the Interpretation of Germline Mutant Alleles (ENIGMA) help in resolving the significance of VUS.[69] Such initiatives have already resulted in a 13% fall in the rate of mutations being called VUS between 2002 and 2013.[70] If the mutation is still reported as VUS, it should not be taken into consideration while arriving at risk assessment in HBOC. Unfortunately, clinicians often want to err on the side of abundant caution and factor in VUS as potentially pathogenic. Caution is advised against this because it may lead to unnecessary prophylactic medication/surgery and/or patient anxiety.[71] [72] [73]


#

Quality of Genetic Testing: The Backbone of Characterizing BRCA1/2 Mutations

All aspects of genetic testing need a robust quality. This includes sample collection and transport; wet lab processing; and bioinformatics. Following established standard operating procedures in genetic testing is extremely important to maintain test quality so as to avoid both false-positive and false-negative reporting (which is true for every aspect of health care management). Following guidelines developed by the American Association of Pathologists' Assistants and the College of American Pathologists for NGS bioinformatics pipelines has been shown to significantly reduce error rates.[66] [74] [75] National accreditation programs and quality assessment programs (e.g., EMQN [European Molecular Genetics Quality Network]) could help meet the goal of optimizing quality.


#

Somatic or Tumor BRCA Testing

DNA extracted from tumor tissue (freshly frozen or from the formalin fixed paraffin embedded [FFPE] blocks) is used to detect somatic mutations. The percentage of tumor cells in the sample processed for mutation analysis can determine the accuracy of the results. If the quality of DNA is compromised, that would also interfere with the test. Further, tissue preservation using formalin induces a chemical crosslinking reaction with nucleotides that results in artefactual sequence alterations and deamination of cytosine nucleotides. These features are of concern when FFPE specimens are used.[76] Use of shorter amplicons, de- crosslinking steps, and treatment with uracil-DNA glycosylase (DNA repair enzyme) are methods that can help reduce the number of sequence artefacts. Their use can improve the quality of extracted DNA.[77] Somatic NGS testing is generally recommended at 500X coverage to avoid false-negative assessment. The bioinformatic pipeline should also be set to take into consideration the percentage of tumor cells in the processed sample.

The ideal somatic mutation testing report should describe[78]:

  • Suitability of tumor sample for tumor content and specific testing method.

  • Number and names of genes tested (if using a multigene panel).

  • Depth of coverage for each gene.

  • Details of mutation (if detected) with HGVS nomenclature.

  • Reference sequence of gene.

  • Interpretation of results with reference to therapy.

When in doubt, somatic mutations identified using NGS may need to be confirmed by Sanger sequencing.

Significance of Somatic (Tumor) BRCA Mutations

In OCs, association with loss of heterozygosity suggests genomic scaring and instability.[73] [74] Sporadic somatic BRCA1/ 2 mutations are seen in up to 33% of BRCA mutations in OC and 4 to 15% of unselected TNBC.[79] [80] [81] [82] Amongst patients with high-grade serous OC (HGSOC), BRCA1/2 germline as well as somatic mutations are frequent (17–25%). In fact, somatic mutations have been reported in 18 to 30% of all patients with BRCA1/2 mutations. It is also interesting to note that 9% of patients with OC showed presence of somatic mutations in homologous recombinant genes (BRCA1/2, BRIP1, CHEK2, and RAD51C).[81]

Clinically, it is important to identify these because presence of somatic mutations is predictive of primary platinum sensitivity and improved overall survival.[83] Somatic BRCA1/2 pathogenic mutations and loss of heterozygosity are also predictive biomarkers for clinical response to PARP inhibitor.[83] [84] [85] [86] Platinum-sensitive relapsed patients with serous OC and positive BRCA mutations have been shown to have the highest chance of benefiting from olaparib (median progression-free survival: 11.2 months in BRCA mutation-positive vs. 7.4 months in wild-type BRCA patients; hazard ratio: 0.54 [95% confidence interval: 0.34–0.85]; p = 0.0075).[87] Based on data from PAOLA-1 study, homologous recombination deficiency (HRD) testing (a composite genomic scar score) has also become an important predictive biomarker for response to PARPi.[88] Some tests developed by other laboratories that can be considered include Myriad MyChoice,[89] loss of heterozygosity, and telomeric allelic imbalance. A word of caution in interpreting their results, since different proprietary algorithms have been incorporated in these tests.[87] Thus, to ensure all eligible patients receive the benefit of PARPi therapy, it is necessary to evaluate all (somatic as well as germline) their BRCA1/2 pathogenic mutations.[90]


#

Somatic or Germline Testing First in Ovarian Cancer?

Somatic mutations testing is not interchangeable with germline testing. Because the bioinformatics pipeline is different, somatic testing is less sensitive. If it is used solely, a significant number (10%) of germline P/LP mutations can be missed.[91] For this reason, most guidelines, including American Society of Clinical Oncology (ASCO), recommend the use of germline testing first, and then if required, somatic testing.[92]

On the other hand, if only germline testing is carried out (on DNA extracted from blood sample), an estimated 5 to 7% of HGSOC cases who may have acquired (somatic) mutations in BRCA1/BRCA2 genes in their tumor will be missed.[50]

So another approach would be to first do tumor testing (to triage cancer genetics referrals) and then offer germline testing to the subset of HGSOC patients in whom a deleterious mutation was detected. For example, somatic P/LP variants seen in tumor specimens are common in some genes with germline implications (e.g., TP53, STK11, PTEN) and may not indicate the need for germline testing unless the clinical/family history is consistent with a P/LP variant in the germline.[51]

Since both approaches have their limitations, current recommendation is to consider parallel testing of somatic and germline panel. This can reduce turnaround time, reduce false negativity but at the cost of double the cost. This is also recommended especially for HGSOC.[52]

European Society for Medical Oncology has recommended either approach, based on physician's choice.[53] We recommend germline testing as the first approach, followed by HRD for those with gBRCA negative.


#
#

Management of HBOC

Risk Management for the Previvor (Unaffected Carrier of Mutation)

  • Lifestyle modifications recommended are:

    • Regular exercise and maintaining a healthy body weight.

    • Limiting alcohol consumption.

    • Avoid hormone-replacement therapy.

    • Encourage breast feeding.

  • Risk reduction surgery: NCCN recommends that BRCA carriers be offered prophylactic bilateral mastectomy.92–94 In both retrospective and prospective observational studies, risk-reducing or prophylactic bilateral mastectomy decreases the incidence of BC by 90% or more in patients who are at risk for hereditary BC, with most studies focusing on BRCA mutation carriers. For BRCA1 carriers, risk-reducing bilateral salpingo-oophorectomy (rrBSO) is recommended for women who have completed child-bearing and should be performed by age 35 to 40 years or individualized on the basis of age of onset of OC in the family.95–97 In BRCA2 carriers, this procedure can be delayed until age 40 to 45 years. rrBSO not only decreases the risk of OC in BRCA mutation carriers, but also decreases the risk of mortality. NCCN does not routinely recommend hysterectomy at the time of rrBSO and indicates that salpingectomy alone is not the standard of care, discouraging it outside a clinical trial.[99]

  • Cancer surveillance: for female BRCA carriers who do not wish to pursue (or would rather delay) surgical risk reduction, BC surveillance should be offered, and OC screening may be performed.[100]

BC screening: the following strategy is recommended by expert groups for women with BRCA pathogenic variants who have not undergone risk-reducing surgery and should be individualized as needed[100] [101]:

  • Breast awareness from 18 years of age.

  • Clinical breast examination every 6 to 12 months is recommended from the age of 25 or 10 years before the youngest BC.

  • Annual screening using magnetic resonance imaging (MRI; days 7 to 15 of the menstrual cycle) should be commenced from age 25 years with the addition of annual mammography with or without tomosynthesis from age 30 years.[78]

  • Age 30 to 75 years: annual mammogram and contrast-enhanced MRI of breast (alternating every 6 months).

  • >75 years: management should be considered on an individual basis.

  • Male BC: breast self-examination training and education starting at 35 years. Clinical breast exam every 12 months starting at 35 years.

OC screening: before rrBSO, 6 monthly transvaginal ultrasound and measure of serum CA-125 may be considered from age 30 years; however, the limited value of these tools as effective screening measures should be communicated to individuals.

  • Pharmacoprevention: use of tamoxifen may be considered; however, the level of evidence is weak.[103] It should be used only for BRCA2 tumors or if the first cancer was estrogen receptor–positive. Its benefit is offset by risk of side effects. The dose (1–20 mg per day) and duration (2–5 years) are also not clear. If raloxifene is used instead of tamoxifen, both efficacy and toxicity are reduced.

  • Prevention of other BRCA-related cancers: no evidence-based data exist. BRCA2 carriers may consider annual skin and eye examination as screening for melanoma, and annual screening for pancreatic cancer with endoscopic ultrasound or MRI/magnetic resonance cholangiopancreatography. There is no consensus when screening should commence; however, age 50 years or 10 years before the earliest diagnosed case in the family would be reasonable ([Table 5]).

  • Reproductive counselling: pathogenic variants in many BC genes, including BRCA, are inherited in an autosomal-dominant pattern, meaning that there is a 50% chance that children of BRCA carriers will have inherited the cancer predisposition variant (assuming that the pathogenic mutation is harbored on only one of the chromosomes of one of the parents). Reproductive counselling of BRCA carriers includes education about prenatal diagnosis and assisted reproduction.[20] One option is preimplantation genetic diagnosis, which is used to analyze embryos, obtained by in vitro fertilization, genetically before their transfer into the uterus.

Table 5

Management recommendations for other genes implicated in hereditary breast cancers

Gene

Breast cancer risk management

Ovarian cancer risk management

Other cancers

ATM

 • Absolute risk 20–40%

 • Consider CEMRI breast from 30–35

 • Potentially increased risk

 • RRSO, insufficient evidence

 • Counsel for autosomal-recessive condition in offspring

BARD1

 • Potentially increased risk

 • RRM insufficient evidence

 • Unknown

BRIP1

 • Unknown

 • Increased risk of ovarian cancer

 • Consider RRSO at age 45–50

CDH1

 • Increased risk of lobular cancer

 • Annual mammography from age of 30 years

 • Discuss option of RRM

 • No increase

 • Gastric cancer: prophylactic total gastrectomy at 20 years or 5 years earlier than the earliest case of HDGC in the family.

CHEK2

 • Increased risk

 • Annual mammogram from age 40 years

 • No increase

 • Colon

NF1

 • Increased risk

 • Annual mammogram from 40 years age

 • RRM insufficient evidence

 • No increase

 • MPNST, GIST

NBN

 • Increased risk

 • Annual mammogram from 40 years age

 • RRM insufficient evidence

 • Unknown

Lynch syndrome genes (MSH2/MSH6/MLH1/PMS2/EPCAM)

 • Unknown or insufficient evidence

 • Except for PMS2, the association is strong. RRSO should be discussed with the patient as evidence is limited.

 • Colon, uterus

PALB2

 • Absolute risk: 41–60%

 • Annual mammogram and breast MRI with contrast at 30 yc,

 • Risk reduction: discuss option of RRM

 • Strength of evidence of association with cancer: strong

 • Absolute risk: 3–5%

 • Management:

 • Risk reduction: consider RRSO at age >45 years

 • Strength of evidence of association with cancer: strong

 • Pancreatic cancer

 • Absolute risk: 5–10%

 • Management: screen P/LP variant carriers with a family history of pancreatic cancer

PTEN

 • Breast awareness from 18 years age

 • CBE every 6–12 months from age of 25 years

 • MRI/mammography from age 30 years or 5–10 years before earlier case in family

 • Endometrial cancer, education, and hysterectomy

 • Annual thyroid USG

 • Colonoscopy every 5 years from age of 35 years

RAD51C

 • Absolute risk: 20–40%

 • Management: annual mammogram and consider breast MRI with contrast starting at age 40 years

 • Strength of evidence of association with cancer: strong

 • Absolute risk: 10–15%

 • Management: risk reduction: recommend RRSO at 45–50 years

 • Strength of evidence of association with cancer: strong

RAD51D

 • Absolute risk: 20–40%

 • Management: annual mammogram and consider breast MRI with contrast starting at age 40 years

 • Strength of evidence of association with cancer: strong

 • Absolute risk: 10–20% 10-12,63,64

 • Management: risk reduction: recommend RRSO at 45–50 years

 • Strength of evidence of association with cancer: strong

STK11

 • Absolute risk: 32–54% •

 • Management: screening: annual mammogram and breast MRI with contrast starting at age 30 years

 • No established association

 • Pancreatic cancer

 • Nonepithelial ovarian cancers

TP53

 • Breast awareness from age of 18 years

 • CBE every 6–12 months from age of 25 years

 • Annual MRI with contrast ages 20–29

 • Annual MRI + mammography ages 30–75

 • RRM to be discussed

 • Annual whole-body MRI

 • Annual brain MRI

 • Colonoscopy and UGIE every 2–5 years from age of 25 years

 • Comprehensive physical and neurological exam\follow Toronto protocol

Abbreviation: MRI, magnetic resonance imaging.



#

Risk Management for Patients

  • Decision for breast-conserving surgery (BCS) versus B/L mastectomy: for a patient with BC, who is technically eligible for BCS, a BRCA mutation is not a contraindication for BCS. Both BCS and mastectomy are equally recommended by ASCO guidelines.[102] Surgical management of the index malignancy (BCT vs. ipsilateral therapeutic mastectomy and contralateral risk-reducing mastectomy [CRRM]) in BRCA1/2 mutation carriers should be discussed, considering the increased risk of contralateral BC (CBC) and possible increased risk of an ipsilateral new primary BC compared with noncarriers. The risks and benefits of both approaches and reconstruction options should be weightage to her age, expectations, wishes, risk acceptance attitude, and body image preferences. For women who have a mutation in a moderate-penetrance BC susceptibility gene, mutation status alone should not determine local therapy decisions for the index tumor or CRRM. BCT should be offered to those for whom BCT is an appropriate treatment option.

  • Risk of OC: rrBSO. Recommendations are the same as those for previvors. There are conflicting data whether rrBSO reduces the risk of BC, with many recent studies not showing any association between rrBSO and BC risk.[96] [97]

  • Risk of CBC [103] [104]: for women with BC who have a BRCA1/2 mutation and who have been treated or are being treated with unilateral mastectomy, CRRM should be offered. Nipple sparing mastectomy is a reasonable option. CRRM is associated with a decreased risk of CBC; there is insufficient evidence for improved survival. The following factors should be considered for assessing risk of CBC and role of risk-reducing mastectomy in BRCA1/2 mutation carriers: age at diagnosis (the strongest predictor of future CBC), family history of BC, overall prognosis from this or other cancers (e.g., ovarian), ability of patient to undergo appropriate breast surveillance (MRI), co-morbidities, and life expectancy.

    BRCA1/2 mutation carriers who do not have bilateral mastectomy should undergo high-risk breast screening of remaining breast tissue with annual mammogram and MRI. Risk-reduction mastectomy and CRRM are not advised routinely to patients with a personal history of advanced OCs, because of the increased chances of recurrence of the index ovarian malignancy.

  • Fertility preservation: in a large analysis including 250 BRCA carriers and 578 controls, it was reported that female BRCA1 carriers had an approximately 33 percent lower anti-mullerian hormone levels relative to controls, although levels in BRCA2 carriers were not diminished. This finding may represent decreased ovarian reserve, and therefore fertility counselling may be appropriate for BRCA1 carriers, if decisions regarding chemotherapy or delayed child-bearing are being considered.[105]


#

Medical Implications of P/LP BRCA Mutations in BC

For patients harboring pathogenic or likely pathogenic BRCA1/2 mutation, PARPi have a role in the management of HER2-negative metastatic stages as well as select high-risk early-stage BCs ([Table 6]).

Table 6

Results from selected PARPi studies in breast and ovarian cancer plus BRCA1/2 mutations

Study treatment (trial)

Indication

Efficacy findings

Breast

 Olaparib monotherapy (300 mg twice per day) vs. standard single agent therapy[93] [106]

 (OLYMPIAD)

Metastatic breast cancer and gBRCA1/2m

ORR: 60% (olaparib) vs. 29% (standard therapy)

Median PFS:

7.0 months (olaparib) vs. 4.2 months (standard therapy; HR:

0.58; 95% CI: 0.43–0.80; p < 0.001)

 Talazoparib monotherapy (1 mg every day) vs. standard single agent therapy[90]

 (EMBRACA)

Metastatic breast cancer and gBRCA1/2m

ORR:

62.6% (talazoparib) vs. 27.2% (standard therapy)

Median PFS: 8.6 months (talazoparib) vs. 5.6 months (standard

therapy; HR: 0.54; 95% CI: 0.41–0.71; p < 0.001)

 Olaparib (300 mg twice per day) vs. placebo[90]

 (OLYMPIA)

High-risk early breast cancer (see the text) and gBRCA1/2m

iDFS at 3 years: 86% (olaparib) vs. 77% (placebo; HR: 0.58; 95% CI: 0.41–0.82; p < 0.001)

OS at 4 years: 89.8% (olaparib) vs. 86.4% (placebo; HR: 0.68; 98.5% CI: 0.47–0.97; p = 0.009)

Ovary

 Olaparib monotherapy (300 mg twice per day) vs. placebo[97] (SOLO 1)

High-grade serous or endometrioid OC, primary peritoneal

cancer, or fallopian tube cancer and g/s BRCA1/2m (FRONTLINE)

PFS at 3 years: 60% (olaparib) vs. 27% (placebo; HR: 0.30; 95% CI:

0.23–0.41; p < 0.001)

 Olaparib monotherapy (300 mg twice per day) vs.

 placebo[94] (SOLO 2)

Platinum-sensitive, relapsed high-grade serous or endometrioid OC, primary peritoneal

cancer, or fallopian tube cancerandgBRCA1/

2m

Median PFS: 19.1 months (olaparib) vs. 5.5 months (placebo; HR:

0.30; 95% CI: 0.22–0.41; p < 0.0001)

 Niraparib monotherapy (300 mg once daily) vs. placebo[95] (NOVA)

Platinum-sensitive relapsed high-grade ovarian cancer gBRCAm cohort

Median PFS: 21 months (niraparib) vs. 5.5 months (placebo; HR: 0.27; 95% CI: 0.17–0.41; p < 0.0001)

 Rucaparib monotherapy (600 mg twice per day)[96] (ARIEL2)

Relapsed high-grade ovarian carcinoma and a g/sBRCA1/2m

ORR: 53.8%; CR: 8.5%; PR: 45.3%; DOR: 9.2 months (95% CI:

6.6–11.6 months)

 Olaparib + bevacizumab vs. placebo + bevacizumab

 (PAOLA-1)

1L maintenance of advanced HGSOC/endometrioid carcinoma patients who have responded to chemotherapy and bevacizumab

mPFS = 22.1 m vs. 16.6 m [PFS (HR): 0.59 (0.49–0.72)]

HRD-positive and BRCA + : PFS (HR) = 0.33 (0.25–0.45)

HRD-positive and BRCA − : PFS (HR) = 0.43(0.28–0.66)

Abbreviations: CI, confidence interval; HR, hazard ratio.


Note: Reference [95] to be changed - Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 2016;375(22):2154–2164.


  • Metastatic/advanced stage setting: olaparib is recommended for patients with germline BRCA mutations and human epidermal growth factor receptor 2–negative BC previously treated with chemotherapy in the neoadjuvant, adjuvant, or metastatic disease setting (based on the Olympiad trial).[107]

Talazoparib is recommended for patients with germline BRCA mutations and human epidermal growth factor receptor 2–negative locally advanced or metastatic BC (based on the EMBRACA trial).[108]

  • Neoadjuvant setting: neoadjuvant platinum chemotherapy is recommended for BRCA-positive patients (based on the GeparSixto and CALGB 40603 trials).[109] [110]

  • Adjuvant setting: one year of adjuvant olaparib is indicated after the completion of (neo)adjuvant chemotherapy and local treatment, including radiation, for patients with high-risk BC (based on the OlympiA trial).[111]

The high-risk patients are[112]:

Post-op TNBC with node-positive disease or an invasive tumor size of at least 2 cm.

Post-op TNBC who have also received neoadjuvant therapy with residual invasive cancer on histopathology (not in pathological complete response).

Hormone-positive BC with at least four involved axillary nodes.

Hormone-positive BC post-neoadjuvant therapy with residual disease and a CPS + EG score of at least 3 (CPS = clinical and pathological stage; EG = estrogen-receptor status and histologic grade).


#

Medical Implications of BRCA in OC

PARPi (olaparib, rucaparib, and niraparib) have been approved in OC for various indications. The broad applications are as below[88] [113] [114] [115] [116]:

  • Frontline setting: as maintenance therapy for advanced epithelial OCs who experienced a response to frontline, platinum-based chemotherapy.

  • Relapsed/recurrent setting: as maintenance therapy for platinum-sensitive relapsed epithelial OCs, irrespective of BRCA status, who has responded to platinum-based therapy rechallenge and who did not receive PARPi maintenance in frontline setting.


#
#

Discussion

These revised/updated consensus guidelines are recommended to be used by health care professionals in their real-world practice (changes in this version of the recommendation guidelines are highlighted in [Table 7] for convenience of the readers). In the absence of prospective randomized data from India, we have not made any attempt to grade or distinguish regarding the strength of the recommendations. [Box 3] shows the summary of the expert group's recommendations, following which is expected to personalize management plan for each patient and ultimately optimize outcome. In the absence of any nationwide genetic testing policy document, our guidelines will serve as its substitute.

Table 7

Key highlights of the major revisions in the current HBOC article as compared to our previous publication (JGO 2020) are as follows:

1. List of non-BRCA genes updated (RAD51C and 1D)

2. Pretest counselling section added

3. Informed consent importance outlined

4. Pedigree chats with sample of the same with commonly used symbols added

5. Implications of germline BRCA and risks of germline BRCA1/2 mutations (U.S. Preventive Services Task Force recommendation added) as well as implications for management of HBOC

6. Details of how to avoid common mistakes made by labs while testing for hereditary cancer panel testing

7. Medical Implications of BRCA in BC section including implications with respect to use of PARPi

8. New, recent Indian references added and updated in [Table 3]

9. Several new genes added and updated in [Table 5]

10. New indications and approval for use of PARPi added in [Table 6]

11. Appropriately, several references updated in the whole manuscript, especially in sections on Somatic/Tumor BRCA testing, BRCA, and PARPi (earlier manuscript had 98 references and the updated one has 121 references)

Box 3

Summary of the ISMPO consensus recommendation statements for HBOC (with voting by authors)

Question

Recommendation (description)

Recommendation (strength)

1. Who should undergo genetic counseling?

All clinicians should assess:

 • Women with a personal or family history of breast, ovarian, tubal, or peritoneal cancer.

 • An ancestry associated BRCA1/2 gene mutations.

and advise genetic counselling and, if indicated, genetic testing

100%

2. Who should undergo genetic testing?

 • Any breast cancer (BC) diagnosed at age <50 years.

 • Any triple-negative BC

 • Any male BC

 • BC at any age and ≥1 close relative (first/second/third degree relative on same side of family) diagnosed with BC, ovarian cancer (OC), prostate, or pancreatic cancer.

 • Any woman with OC

96%

3. Who can perform genetic counselling?

Genetic counsellors and other medical professionals (medical/surgical/radiation oncologists/breast surgeons) knowledgeable in genetic testing can provide patient education and counselling and make recommendations regarding genetic testing and arrange testing

100%

4. What points should be included in pretest counselling?

 • Rapport building,

 • Elicitation of need and comprehension levels

 • Medical history and pedigree evaluation

 • Decide the best test candidate to test first

 • Genetic testing recommendations

 • Implications of genetic testing: benefits/harms

 • Variants of unknown significance

 • Financial considerations

 • Risk reduction options

100%

5. What genetic test should be offered?

 • Single-site mutation testing in families with a known mutation

 • For unknown mutation:

   - Essential: BRCA1/2 sequencing by next-generation sequencing plus multiplex ligation probe amplification (MLPA; BRCA1/2) for large genomic rearrangements (LGRs)

   - Desirable: multigene panel testing (a representative model panel should include BRCA1, BRCA2, p53, PTEN, CDH1, PALB2, CHEK2, ATM, RAD51C, STK11, RAD51D, BRIP1, MLH1, MSH2, MSH6, and PMS2) + MLPA (BRCA1/2) for LGRs

100%

6. What risk-reduction approaches should be offered to affected individuals?

 • Risk management for future cancers:

  Contralateral prophylactic mastectomy:

   ➢ Risk-reduction mastectomy should be offered to patients with a previous history of BC who carry a germline genetic mutation in BRCA1/2

   ➢ Risk-reducing bilateral salpingo-oophorectomy (rrBSO): for BRCA1 carriers, rrBSO is recommended for women who have completed childbearing, and should be performed by age 35 to 40 years. In BRCA2 carriers, one can consider delaying this procedure until age 40 to 45

 • Advanced OC with BRCA mutation: prophylactic bilateral mastectomy is not considered in these cases as the risk of death from the primary malignancy is high over the next 5 years. In these cases, nonsurgical measures and surveillance only are used for any new primary malignancy in breasts

100%

7. What risk-reduction approaches should be offered to unaffected mutation carriers?

BRCA1/2:

 • Lifestyle modifications: regular exercise, maintaining healthy body weight, limiting alcohol consumption.

 • Avoid hormone replacement therapy, encourage breast feeding.

 • Breast cancer: BRCA carriers should be offered prophylactic bilateral mastectomy; however, the final decision is based on personal preference, given that effective screening is available.

 • Bilateral salpingo-oophorectomy: for BRCA carriers, risk-reducing bilateral salpingo-oophorectomy is recommended for women who have completed childbearing, and should be performed by age 35 to 40 years. In BRCA2 carriers, one can consider delaying this procedure until age 40 to 45 years.

 • Cancer surveillance: for female BRCA carriers who do not wish to pursue (or would rather delay) surgical risk reduction, BC surveillance should be offered, and OC screening may be performed

 • Breast cancer screening:

   ➢ Breast awareness from age 18 years IIA

   ➢ Clinical breast examination (CBE) every 6–12 months is recommended from the age of 25 years or 10 years before the youngest BC VC

   ➢ Annual screening MRI (days 7–15 of menstrual cycle) should be commenced from age 25 years with the addition of annual mammography from age 30 years

  • OC screening:

   ➢ Concurrent transvaginal ultrasound (preferably days 1–10 of menstrual cycle) and CA-125 (best performed after day 5 of menstrual cycle) every 6 months beginning at age 30 years

   ➢ Before risk-reducing bilateral salpingo-oophorectomy, 6 monthly transvaginal ultrasound and measures of serum CA-125 may be considered from age 30 years; however, the limited value of these tools as an effective screening measure should be communicated to individuals

   ➢ Chemoprevention: use of tamoxifen may be considered; however, the level of evidence is weak; use tamoxifen only for BRCA2 tumors or if the first cancer was estrogen receptor-positive

   ➢ Surveillance in male previvors: There are no proven risk-reducing surgical options for men

   - Monthly breast self-examination starting at age 35 years

   - Clinical breast examination every 12 months starting at age 35 years

   - Prostate cancer screening starting at age 45 years for BRCA2 carriers and consideration of prostate screening for BRCA1 carriers also at age 45 years

100%

8. When should poly (ADP-ribose) polymerase inhibitors be used?

 ➢ Olaparib, niraparib, and rucaparib are indicated for maintenance treatment in adults with recurrent epithelial OC who are in complete response (CR) or partial response (PR) after platinum-based chemotherapy (irrespective of BRCA status)

 ➢ In g/s P/LP BRCA-mutated OC, olaparib should be used as maintenance after a CR/PR to first-line chemotherapy and cytoreductive surgery. Niraparib is indicated in this setting irrespective of BRCA status.

 ➢ 1L maintenance treatment for advanced ovarian cancer in combination with bevacizumab after CR/PR to 1L platinum-based chemotherapy which is HRD+

 ➢ Talazoparib is indicated for adults with deleterious or suspected gBRCA-mutated, human epidermal growth factor receptor 2–negative locally advanced or metastatic BC

 ➢ Olaparib is indicated for adjuvant treatment of P/LP gBRCA HER2-negative high-risk early breast cancer in adults previously treated with NACT or adjuvant CT

 ➢ Olaparib is indicated for P/LP gBRCA HER2-negative metastatic breast cancer patients who have received chemotherapy in neoadjuvant/adjuvant/metastatic setting. Hormone-positive breast cancer should have been treated with prior endocrine therapy or be considered inappropriate for endocrine therapy.

96%

Abbreviation: CT, computed tomography.



#

Conclusion

HBOC is a complex syndrome with additional insights constantly being generated from real-world experiences combined with data science developments in global publicly available databases.[117] The community oncologists in India, SAARC region, and other low- and middle-income countries are in the need of guidelines that can be impropriated easily into their clinical practice to optimize genetic counselling, molecular testing, and management of patients with HBOC. This is because the implications are far-reaching in our socioeconomic milieu. This consensus statement provides a clear picture of why to test, whom to test, how to test, when to test, which test to use, how to interpret the results, and how to discuss options with the patient and families at risk.[118] [119] [120] [121]

HBOC ISMPO Guidelines (2023)


#
#

Conflict of Interest

None declared.

Acknowledgements

Our sincere thanks to Late Prof. Dr. G.S. Bhhattacharyya for his seminal role in initiating this project and leading it in the first guidelines published in 2020.

Patient Consent

Patient consent not required.


  • References

  • 1 Bhardwaj PV, Dulala R, Rajappa S, Loke C. Breast cancer in India: screening, detection, and management. Hematol Oncol Clin North Am 2024; 38 (01) 123-135
  • 2 Mohanty SK, Wadasadawala T, Sen S, Khan PK. Socio-economic variations of breast cancer treatment and discontinuation: a study from a public tertiary cancer hospital in Mumbai, India. BMC Womens Health 2023; 23 (01) 113
  • 3 Waghela BN, Pandit RJ, Puvar A. et al. Identification of novel exonic variants contributing to hereditary breast and ovarian cancer in west Indian population. Gene 2023; 852: 147070
  • 4 American College of Obstetricians and Gynecologists. ACOG Committee on Practice Bulletins—Gynecology, ACOG Committee on Genetics. et al. ACOG Practice Bulletin No. 103: Hereditary breast and ovarian cancer syndrome. Obstet Gynecol 2009; 113: 957-966 PubMed
  • 5 Honrado E, Benítez J, Palacios J. The molecular pathology of hereditary breast cancer: genetic testing and therapeutic implications. Mod Pathol 2005; 18 (10) 1305-1320
  • 6 Ford D, Easton DF, Stratton M. et al; The Breast Cancer Linkage Consortium. Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. Am J Hum Genet 1998; 62 (03) 676-689
  • 7 Gorodetska I, Kozeretska I, Dubrovska A. BRCA genes: the role in genome stability, cancer stemness and therapy resistance. J Cancer 2019; 10 (09) 2109-2127
  • 8 Struewing JP, Hartge P, Wacholder S. et al. The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 1997; 336 (20) 1401-1408
  • 9 Sarin R. A decade of discovery of BRCA1 and BRCA2: are we turning the tide against hereditary breast cancers?. J Cancer Res Ther 2006; 2 (04) 157-158
  • 10 Joshi S, Murali-Nanavati S, Shylasree TS. et al. Synchronous and metachronous breast and ovarian cancers: experience from a single tertiary care cancer centre in India. Indian J Surg Oncol 2023; 14 (04) 809-821
  • 11 Marsh D, Zori R. Genetic insights into familial cancers– update and recent discoveries. Cancer Lett 2002; 181 (02) 125-164
  • 12 Rhiem K, Schmutzler R. Impact of prophylactic mastectomy in BRCA1/2 mutation carriers. Breast Care (Basel) 2014; 9 (06) 385-389
  • 13 Claus EB, Risch N, Thompson WD. Autosomal dominant inheritance of early-onset breast cancer. Implications for risk prediction. Cancer 1994; 73 (03) 643-651
  • 14 Krivokuca A, Mihajlovic M, Susnjar S. et al. Mutational profile of hereditary breast and ovarian cancer - establishing genetic testing guidelines in a developing country. Curr Probl Cancer 2022; 46 (01) 100767
  • 15 Peto J, Collins N, Barfoot R. et al. Prevalence of BRCA1 and BRCA2 gene mutations in patients with early-onset breast cancer. J Natl Cancer Inst 1999; 91 (11) 943-949
  • 16 Rajagopal T, Seshachalam A, Jothi A. et al. Analysis of pathogenic variants in BRCA1 and BRCA2 genes using next-generation sequencing in women with triple negative breast cancer from South India. Mol Biol Rep 2022; 49 (04) 3025-3032
  • 17 Forbes C, Fayter D, de Kock S, Quek RG. A systematic review of international guidelines and recommendations for the genetic screening, diagnosis, genetic counseling, and treatment of BRCA-mutated breast cancer. Cancer Manag Res 2019; 11: 2321-2337
  • 18 Kang PC, Phuah SY, Sivanandan K. et al. Recurrent mutation testing of BRCA1 and BRCA2 in Asian breast cancer patients identify carriers in those with presumed low risk by family history. Breast Cancer Res Treat 144: 635-642 2014;
  • 19 Malhotra H, Kowtal P, Mehra N. et al. Genetic counseling, testing, and management of HBOC in India: an expert consensus document from Indian Society of Medical and Pediatric Oncology. JCO Glob Oncol 2020; 6: 991-1008
  • 20 Parikh PM, Aggarwal S, Biswas G. et al. Practical clinical consensus guidelines for the management of cancer associated anemia in low- and middle-income countries. South Asian J Cancer 2023; 12 (02) 93-99
  • 21 Phadke SR, Pandey A, Puri RD, Patil SJ. Genetic counseling: the impact in Indian milieu. Indian J Pediatr 2004; 71 (12) 1079-1082
  • 22 National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Genetic/familial high-risk assessment—Breast, Ovarian, and pancreatic (version 1.2020). Accessed July 11, 2024 at: https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf
  • 23 Riley BD, Culver JO, Skrzynia C. et al. Essential elements of genetic cancer risk assessment, counseling, and testing: updated recommendations of the National Society of Genetic Counselors. J Genet Couns 2012; 21 (02) 151-161
  • 24 Allen CG, Roberts M, Guan Y. Exploring predictors of genetic counseling and testing for hereditary breast and ovarian cancer: findings from the 2015 U.S. National Health Interview Survey. J Pers Med 2019; 9 (02) 26
  • 25 Fallowfield L, Jenkins V. Effective communication skills are the key to good cancer care. Eur J Cancer 1999; 35 (11) 1592-1597
  • 26 Robson ME, Bradbury AR, Arun B. et al. American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol 2015; 33 (31) 3660-3667
  • 27 National Pathology Accreditation Advisory Council. Classification of Human Genetic Testing. Canberra, ACT, Australia, Laboratory Accreditation Standards and Guidelines for Nucleic Acid Detection and Analysis; 2007
  • 28 Rantanen E, Hietala M, Kristoffersson U. et al. Regulations and practices of genetic counselling in 38 European countries: the perspective of national representatives. Eur J Hum Genet 2008; 16 (10) 1208-1216
  • 29 Darooei M, Poornima S, Salma BU. et al. Pedigree and BRCA gene analysis in breast cancer patients to identify hereditary breast and ovarian cancer syndrome to prevent morbidity and mortality of disease in Indian population. Tumour Biol 2017; 39 (02) 1010428317694303
  • 30 Rich EC, Burke W, Heaton CJ. et al. Reconsidering the family history in primary care. J Gen Intern Med 2004; 19 (03) 273-280
  • 31 Yip CH, Evans DG, Agarwal G. et al. Global disparities in breast cancer genetics testing, counselling and management. World J Surg 2019; 43 (05) 1264-1270
  • 32 Wood ME, Kadlubek P, Pham TH. et al. Quality of cancer family history and referral for genetic counseling and testing among oncology practices: a pilot test of quality measures as part of the American Society of Clinical Oncology Quality Oncology Practice Initiative. J Clin Oncol 2014; 32 (08) 824-829
  • 33 Brédart A, Kop JL, De Pauw A. et al. Effect on perceived control and psychological distress of genetic knowledge in women with breast cancer receiving a BRCA1/2 test result. Breast 2017; 31: 121-127
  • 34 Mancini J, Noguès C, Adenis C. et al. Impact of an information booklet on satisfaction and decision-making about BRCA genetic testing. Eur J Cancer 2006; 42 (07) 871-881
  • 35 Quinn VF, Meiser B, Kirk J. et al. Streamlined genetic education is effective in preparing women newly diagnosed with breast cancer for decision making about treatment-focused genetic testing: a randomized controlled noninferiority trial. Genet Med 2017; 19 (04) 448-456
  • 36 Randall J, Butow P, Kirk J, Tucker K. Psychological impact of genetic counselling and testing in women previously diagnosed with breast cancer. Intern Med J 2001; 31 (07) 397-405
  • 37 National Cancer Institute. Susceptibility genes. Accessed July 11, 2024 at: https://www.cancer.gov/types/breast/hp/breast-ovarian-genetics-pdq
  • 38 Kowalewski A, Szylberg Ł, Saganek M, Napiontek W, Antosik P, Grzanka D. Emerging strategies in BRCA-positive pancreatic cancer. J Cancer Res Clin Oncol 2018; 144 (08) 1503-1507
  • 39 Singh J, Thota N, Singh S. et al. Screening of over 1000 Indian patients with breast and/or ovarian cancer with a multi-gene panel: prevalence of BRCA1/2 and non-BRCA mutations. Breast Cancer Res Treat 2018; 170 (01) 189-196
  • 40 Agarwal G, Pradeep PV, Aggarwal V, Yip CH, Cheung PS. Spectrum of breast cancer in Asian women. World J Surg 2007; 31 (05) 1031-1040
  • 41 Sharma-Oates A, Shaaban AM, Tomlinson I, Wynne L, Cazier JB, Sundar S. Heterogeneity of germline variants in high risk breast and ovarian cancer susceptibility genes in India. Precis Clin Med 2018; 1 (02) 75-87
  • 42 Thirthagiri E, Lee SY, Kang P. et al. Evaluation of BRCA1 and BRCA2 mutations and risk-prediction models in a typical Asian country (Malaysia) with a relatively low incidence of breast cancer. Breast Cancer Res 2008; 10 (04) R59
  • 43 Gupta S, Rajappa S, Advani S. et al. Prevalence of BRCA1 and BRCA2 mutations among patients with ovarian, primary peritoneal, and fallopian tube cancer in India: a multicenter cross-sectional study. JCO Glob Oncol 2021; 7: 849-861
  • 44 Mittal A, Deo SVS, Gogia A. et al. Profile of pathogenic mutations and evaluation of germline genetic testing criteria in consecutive breast cancer patients treated at a North Indian tertiary care center. Ann Surg Oncol 2022; 29 (02) 1423-1432
  • 45 Pramanik R, Upadhyay A, Khurana S. et al. Comprehensive germline genomic profiling of patients with ovarian cancer: a cross-sectional study. Indian J Med Paediatr Oncol 2022; 43 (04) 361-368
  • 46 Kadri MSN, Patel KM, Bhargava PA. et al. Mutational landscape for Indian hereditary breast and ovarian cancer cohort suggests need for identifying population specific genes and biomarkers for screening. Front Oncol 2021; 10: 568786
  • 47 Chheda P, Pande S, Dama T. et al. Spectrum of germline BRCA mutations in hereditary breast and ovarian cancer syndrome in Indian population: A central reference laboratory experience. Cancer Res Stat Treat 2020; 3: 32-41
  • 48 Mehta A, Vasudevan S, Sharma SK. et al. Germline BRCA1 and BRCA2 deleterious mutations and variants of unknown clinical significance associated with breast/ovarian cancer: a report from North India. Cancer Manag Res 2018; 10: 6505-6516
  • 49 Suryavanshi M, Kumar D, Panigrahi MK, Chowdhary M, Mehta A. Detection of false positive mutations in BRCA gene by next generation sequencing. Fam Cancer 2017; 16 (03) 311-317
  • 50 Stewart MD, Merino Vega D, Arend RC. et al. Homologous recombination deficiency: concepts, definitions, and assays. Oncologist 2022; 27 (03) 167-174
  • 51 Bekos C, Grimm C, Kranawetter M. et al. Reliability of tumor testing compared to germline testing for detecting BRCA1 and BRCA2 mutations in patients with epithelial ovarian cancer. J Pers Med 2021; 11 (07) 593
  • 52 Chandrsekaran D, Sobocan M, Blyuss O. et al. Implementation of multigene germline and parallel somatic genetic testing in epithelial ovarian cancer: SIGNPOST study. Cancers (Basel) 2021; 13: 4344
  • 53 Miller RE, Leary A, Scott CL. et al. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann Oncol 2020; 31 (12) 1606-1622
  • 54 Owens DK, Davidson KW, Krist AH. et al; US Preventive Services Task Force. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US Preventive Services Task Force recommendation statement. JAMA 2019; 322 (07) 652-665
  • 55 Vaidyanathan K, Lakhotia S, Ravishankar HM, Tabassum U, Mukherjee G, Somasundaram K. BRCA1 and BRCA2 germline mutation analysis among Indian women from south India: identification of four novel mutations and high-frequency occurrence of 185delAG mutation. J Biosci 2009; 34 (03) 415-422
  • 56 McAlarnen L, Stearns K, Uyar D. Challenges of genomic testing for hereditary breast and ovarian cancers. Appl Clin Genet 2021; 14: 1-9
  • 57 Price KS, Svenson A, King E, Ready K, Lazarin GA. Inherited cancer in the age of next-generation sequencing. Biol Res Nurs 2018; 20 (02) 192-204
  • 58 Ruiz de Garibay G, Gutiérrez-Enríquez S, Garre P. et al. Characterization of four novel BRCA2 large genomic rearrangements in Spanish breast/ovarian cancer families: review of the literature, and reevaluation of the genetic mechanisms involved in their origin. Breast Cancer Res Treat 2012; 133 (01) 273-283
  • 59 Reddy RRS, Ramanujam MV. High throughput sequencing-based approaches for gene expression analysis. Methods Mol Biol 2018; 1783: 299-323
  • 60 Avoiding the common mistakes labs make when launching hereditary cancer panel tests Jeanette McCarthy. Accessed July 11, 2024 at: www.fabricgenomics.com
  • 61 Feliubadaló L, Lopez-Doriga A, Castellsagué E. et al. Next-generation sequencing meets genetic diagnostics: development of a comprehensive workflow for the analysis of BRCA1 and BRCA2 genes. Eur J Hum Genet 2013; 21 (08) 864-870
  • 62 Tucker K. Germline genetic testing: why it matters and where we are failing. Gynaecologic Oncology Consult 2022 . Accessed November 13, 2023 at: https://www.mdedge.com/obgyn/article/258782/gynecologic-cancer/germline-genetic-testing-why-it-matters-and-where-we-are
  • 63 Carvalho CM, Braga LDC, Silva LM, Chami AM, Silva Filho ALD. Germline mutations landscape in a cohort of the state of Minas Gerais, Brazil, in patients who underwent genetic counseling for gynecological and breast cancer. Rev Bras Ginecol Obstet 2023; 45 (02) 74-81
  • 64 Wallace AJ. New challenges for BRCA testing: a view from the diagnostic laboratory. Eur J Hum Genet 2016; 24 (Suppl. 01) S10-S18
  • 65 Jennings LJ, Arcila ME, Corless C. et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn 2017; 19 (03) 341-365
  • 66 Richards S, Aziz N, Bale S. et al; ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 2015; 17 (05) 405-424
  • 67 Roy S, Coldren C, Karunamurthy A. et al. Standards and guidelines for validating next-generation sequencing bioinformatics pipelines: a joint recommendation of the Association for Molecular Pathology and the College of American Pathologists. J Mol Diagn 2018; 20 (01) 4-27
  • 68 Do H, Wong SQ, Li J, Dobrovic A. Reducing sequence artifacts in amplicon-based massively parallel sequencing of formalin-fixed paraffin-embedded DNA by enzymatic depletion of uracil-containing templates. Clin Chem 2013; 59 (09) 1376-1383
  • 69 Hoppe MM, Sundar R, Tan DSP, Jeyasekharan AD. Biomarkers for homologous recombination deficiency in cancer. J Natl Cancer Inst 2018; 110 (07) 704-713
  • 70 Plon SE, Eccles DM, Easton D. et al; IARC Unclassified Genetic Variants Working Group. Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results. Hum Mutat 2008; 29 (11) 1282-1291
  • 71 Meijers-Heijboer H, van Geel B, van Putten WL. et al. Breast cancer after prophylactic bilateral mastectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 2001; 345 (03) 159-164
  • 72 Domchek SM, Friebel TM, Singer CF. et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA 2010; 304 (09) 967-975
  • 73 Kauff ND, Satagopan JM, Robson ME. et al. Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 2002; 346 (21) 1609-1615
  • 74 Zuntini R, Ferrari S, Bonora E. et al. Dealing with BRCA1/2 unclassified variants in a cancer genetics clinic: does cosegregation analysis help?. Front Genet 2018; 9: 378
  • 75 Neff RT, Senter L, Salani R. BRCA mutation in ovarian cancer: testing, implications and treatment considerations. Ther Adv Med Oncol 2017; 9 (08) 519-531
  • 76 Nagahashi M, Shimada Y, Ichikawa H. et al. Formalin-fixed paraffin-embedded sample conditions for deep next generation sequencing. J Surg Res 2017; 220: 125-132
  • 77 Cazzato G, Caporusso C, Arezzo F. et al. Formalin-fixed and paraffin-embedded samples for next generation sequencing: problems and solutions. Genes (Basel) 2021; 12 (10) 1472
  • 78 Moore DA, Balbi K, Ingham A, Arkenau HT, Bennett P. Analysis of a large cohort of non-small cell lung cancers submitted for somatic variant analysis demonstrates that targeted next-generation sequencing is fit for purpose as a molecular diagnostic assay in routine practice. J Clin Pathol 2018; 71 (11) 1001-1006
  • 79 Rajkumar T, Meenakumari B, Mani S, Sridevi V, Sundersingh S. Targeted resequencing of 30 genes improves the detection of deleterious mutations in South Indian women with breast and/or ovarian cancers. Asian Pac J Cancer Prev 2015; 16 (13) 5211-5217
  • 80 Borg A, Haile RW, Malone KE. et al. Characterization of BRCA1 and BRCA2 deleterious mutations and variants of unknown clinical significance in unilateral and bilateral breast cancer: the WECARE study. Hum Mutat 2010; 31 (03) E1200-E1240
  • 81 Exchange BRCA. . Summary view. Accessed July 11, 2024 at: https://brcaexchange.org/
  • 82 Lindor NM, Goldgar DE, Tavtigian SV, Plon SE, Couch FJ. BRCA1/2 sequence variants of uncertain significance: a primer for providers to assist in discussions and in medical management. Oncologist 2013; 18 (05) 518-524
  • 83 Ledermann J, Harter P, Gourley C. Correction to Lancet Oncol 2014; 15: 856. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 2015; 16 (04) e158
  • 84 Hennessy BT, Timms KM, Carey MS. et al. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol 2010; 28 (22) 3570-3576
  • 85 Ledermann J, Harter P, Gourley C. et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 2014; 15 (08) 852-861
  • 86 Sandhu SK, Schelman WR, Wilding G. et al. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol 2013; 14 (09) 882-892
  • 87 Agarwal A, Baghmar S, Dodagoudar C. et al. PARP inhibitor in platinum-resistant ovarian cancer: single-center real-world experience. JCO Glob Oncol 2021; 7: 506-511
  • 88 Vergote I, Ray-Coquard I, Anderson DM. et al. Population-adjusted indirect treatment comparison of the SOLO1 and PAOLA-1/ENGOT-ov25 trials evaluating maintenance olaparib or bevacizumab or the combination of both in newly diagnosed, advanced BRCA-mutated ovarian cancer. Eur J Cancer 2021; 157: 415-423
  • 89 Kanjanapan Y, Lheureux S, Oza AM. Niraparib for the treatment of ovarian cancer. Expert Opin Pharmacother 2017; 18 (06) 631-640
  • 90 Vendrell JA, Ban IO, Solassol I. et al. Differential sensitivity of germline and somatic BRCA variants to PARP inhibitor in high-grade serous ovarian cancer. Int J Mol Sci 2023; 24 (18) 14181
  • 91 Terraf P, Pareja F, Brown DN. et al. Comprehensive assessment of germline pathogenic variant detection in tumor-only sequencing. Ann Oncol 2022; 33 (04) 426-433
  • 92 Tew WP, Lacchetti C, Ellis A. et al. PARP inhibitors in the management of ovarian cancer: ASCO guideline. J Clin Oncol 2020; 38 (30) 3468-3493
  • 93 Rebbeck TR, Lynch HT, Neuhausen SL. et al; Prevention and Observation of Surgical End Points Study Group. Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 2002; 346 (21) 1616-1622
  • 94 Carbine NE, Lostumbo L, Wallace J, Ko H. Risk-reducing mastectomy for the prevention of primary breast cancer. Cochrane Database Syst Rev 2018; 4 (04) CD002748
  • 95 Lostumbo L, Carbine NE, Wallace J. Prophylactic mastectomy for the prevention of breast cancer. Cochrane Database Syst Rev 2010; CD002748 (11) CD002748
  • 96 Heemskerk-Gerritsen BA, Seynaeve C, van Asperen CJ. et al; Hereditary Breast and Ovarian Cancer Research Group Netherlands. Breast cancer risk after salpingo-oophorectomy in healthy BRCA1/2 mutation carriers: revisiting the evidence for risk reduction. J Natl Cancer Inst 2015; 107 (05) djv033
  • 97 Kotsopoulos J, Huzarski T, Gronwald J. et al; Hereditary Breast Cancer Clinical Study Group. Bilateral oophorectomy and breast cancer risk in BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 2016; 109 (01) djw177
  • 98 Terry MB, Daly MB, Phillips KA. et al. Risk-reducing oophorectomy and breast cancer risk across the spectrum of familial risk. J Natl Cancer Inst 2019; 111 (03) 331-334
  • 99 NCCN. Accessed July 11, 2024 at: https://www.nccn.org/guidelines/guidelines-detail?category=2&id=1420
  • 100 Saslow D, Boetes C, Burke W. et al; American Cancer Society Breast Cancer Advisory Group. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin 2007; 57 (02) 75-89
  • 101 King MC, Wieand S, Hale K. et al; National Surgical Adjuvant Breast and Bowel Project. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA 2001; 286 (18) 2251-2256
  • 102 Tung NM, Boughey JC, Pierce LJ. et al. Management of hereditary breast cancer: American Society of Clinical Oncology, American Society for Radiation Oncology, and Society of Surgical Oncology Guideline. J Clin Oncol 2020; 38 (18) 2080-2106
  • 103 Pesce C, Liederbach E, Wang C, Lapin B, Winchester DJ, Yao K. Contralateral prophylactic mastectomy provides no survival benefit in young women with estrogen receptor-negative breast cancer. Ann Surg Oncol 2014; 21 (10) 3231-3239
  • 104 Wong SM, Freedman RA, Sagara Y, Aydogan F, Barry WT, Golshan M. Growing use of contralateral prophylactic mastectomy despite no improvement inlong term survival for invasive breast cancer. Ann Surg 2017; 265 (03) 581-589
  • 105 Son KA, Lee DY, Choi D. Association of BRCA mutations and anti-müllerian hormone level in young breast cancer patients. Front Endocrinol (Lausanne) 2019; 10: 235
  • 106 Rădoi VE, Țurcan M, Maioru OV. et al. Homologous recombination deficiency score determined by genomic instability in a Romanian cohort. Diagnostics (Basel) 2023; 13 (11) 1896
  • 107 Robson ME, Tung N, Conte P. et al. OlympiAD final overall survival and tolerability results: Olaparib versus chemotherapy treatment of physician's choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol 2019; 30 (04) 558-566
  • 108 Litton JK, Hurvitz SA, Mina LA. et al. Talazoparib versus chemotherapy in patients with germline BRCA1/2-mutated HER2-negative advanced breast cancer: final overall survival results from the EMBRACA trial. Ann Oncol 2020; 31 (11) 1526-1535
  • 109 von Minckwitz G, Schneeweiss A, Loibl S. et al. Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): a randomised phase 2 trial. Lancet Oncol 2014; 15 (07) 747-756
  • 110 Sikov WM, Berry DA, Perou CM. et al. Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance). J Clin Oncol 2015; 33 (01) 13-21
  • 111 Tutt ANJ, Garber JE, Kaufman B. et al; OlympiA Clinical Trial Steering Committee and Investigators. Adjuvant Olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med 2021; 384 (25) 2394-2405
  • 112 Aschenbrenner DS. New adjuvant treatment for high-risk early breast cancer. Am J Nurs 2022; 122 (07) 26
  • 113 DiSilvestro P, Banerjee S, Colombo N. et al; SOLO1 Investigators. Overall survival with maintenance olaparib at a 7-year follow-up in patients with newly diagnosed advanced ovarian cancer and a BRCA mutation: the SOLO1/GOG 3004 trial. J Clin Oncol 2023; 41 (03) 609-617
  • 114 Pujade-Lauraine E, Ledermann JA, Selle F. et al; SOLO2/ENGOT-Ov21 investigators. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18 (09) 1274-1284
  • 115 Fabbro M, Moore KN, Dørum A. et al. Efficacy and safety of niraparib as maintenance treatment in older patients (≥ 70 years) with recurrent ovarian cancer: Results from the ENGOT-OV16/NOVA trial. Gynecol Oncol 2019; 152 (03) 560-567
  • 116 Oza AM, Tinker AV, Oaknin A. et al. Antitumor activity and safety of the PARP inhibitor rucaparib in patients with high-grade ovarian carcinoma and a germline or somatic BRCA1 or BRCA2 mutation: Integrated analysis of data from Study 10 and ARIEL2. Gynecol Oncol 2017; 147 (02) 267-275
  • 117 Moore K, Colombo N, Scambia G. et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 2018; 379 (26) 2495-2505
  • 118 Valarmathi MT, A A, Deo SS, Shukla NK, Das SN. BRCA1 germline mutations in Indian familial breast cancer. Hum Mutat 2003; 21 (01) 98-99
  • 119 Saxena S, Chakraborty A, Kaushal M. et al. Contribution of germline BRCA1 and BRCA2 sequence alterations to breast cancer in Northern India. BMC Med Genet 2006; 7: 75
  • 120 Syamala V, Sreeja L, Syamala VS. et al. Novel germline mutations in BRCA2 gene among 96 hereditary breast and breast-ovarian cancer families from Kerala, South India. J Cancer Res Clin Oncol 2007; 133 (11) 867-874
  • 121 Soumittra N, Meenakumari B, Parija T. et al. Molecular genetics analysis of hereditary breast and ovarian cancer patients in India. Hered Cancer Clin Pract 2009; 7 (01) 13

Address for correspondence

Hemant Malhotra, MD, FRCP, FRCP, FACP, ECMO, FNAMS, FUICC, FICP, FIMSA
Sri Ram Cancer Center, Mahatma Gandhi University of Medical Sciences and Technology
Sitapura, Jaipur 302022, Rajasthan
India   

Publikationsverlauf

Artikel online veröffentlicht:
03. März 2025

© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

  • References

  • 1 Bhardwaj PV, Dulala R, Rajappa S, Loke C. Breast cancer in India: screening, detection, and management. Hematol Oncol Clin North Am 2024; 38 (01) 123-135
  • 2 Mohanty SK, Wadasadawala T, Sen S, Khan PK. Socio-economic variations of breast cancer treatment and discontinuation: a study from a public tertiary cancer hospital in Mumbai, India. BMC Womens Health 2023; 23 (01) 113
  • 3 Waghela BN, Pandit RJ, Puvar A. et al. Identification of novel exonic variants contributing to hereditary breast and ovarian cancer in west Indian population. Gene 2023; 852: 147070
  • 4 American College of Obstetricians and Gynecologists. ACOG Committee on Practice Bulletins—Gynecology, ACOG Committee on Genetics. et al. ACOG Practice Bulletin No. 103: Hereditary breast and ovarian cancer syndrome. Obstet Gynecol 2009; 113: 957-966 PubMed
  • 5 Honrado E, Benítez J, Palacios J. The molecular pathology of hereditary breast cancer: genetic testing and therapeutic implications. Mod Pathol 2005; 18 (10) 1305-1320
  • 6 Ford D, Easton DF, Stratton M. et al; The Breast Cancer Linkage Consortium. Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. Am J Hum Genet 1998; 62 (03) 676-689
  • 7 Gorodetska I, Kozeretska I, Dubrovska A. BRCA genes: the role in genome stability, cancer stemness and therapy resistance. J Cancer 2019; 10 (09) 2109-2127
  • 8 Struewing JP, Hartge P, Wacholder S. et al. The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 1997; 336 (20) 1401-1408
  • 9 Sarin R. A decade of discovery of BRCA1 and BRCA2: are we turning the tide against hereditary breast cancers?. J Cancer Res Ther 2006; 2 (04) 157-158
  • 10 Joshi S, Murali-Nanavati S, Shylasree TS. et al. Synchronous and metachronous breast and ovarian cancers: experience from a single tertiary care cancer centre in India. Indian J Surg Oncol 2023; 14 (04) 809-821
  • 11 Marsh D, Zori R. Genetic insights into familial cancers– update and recent discoveries. Cancer Lett 2002; 181 (02) 125-164
  • 12 Rhiem K, Schmutzler R. Impact of prophylactic mastectomy in BRCA1/2 mutation carriers. Breast Care (Basel) 2014; 9 (06) 385-389
  • 13 Claus EB, Risch N, Thompson WD. Autosomal dominant inheritance of early-onset breast cancer. Implications for risk prediction. Cancer 1994; 73 (03) 643-651
  • 14 Krivokuca A, Mihajlovic M, Susnjar S. et al. Mutational profile of hereditary breast and ovarian cancer - establishing genetic testing guidelines in a developing country. Curr Probl Cancer 2022; 46 (01) 100767
  • 15 Peto J, Collins N, Barfoot R. et al. Prevalence of BRCA1 and BRCA2 gene mutations in patients with early-onset breast cancer. J Natl Cancer Inst 1999; 91 (11) 943-949
  • 16 Rajagopal T, Seshachalam A, Jothi A. et al. Analysis of pathogenic variants in BRCA1 and BRCA2 genes using next-generation sequencing in women with triple negative breast cancer from South India. Mol Biol Rep 2022; 49 (04) 3025-3032
  • 17 Forbes C, Fayter D, de Kock S, Quek RG. A systematic review of international guidelines and recommendations for the genetic screening, diagnosis, genetic counseling, and treatment of BRCA-mutated breast cancer. Cancer Manag Res 2019; 11: 2321-2337
  • 18 Kang PC, Phuah SY, Sivanandan K. et al. Recurrent mutation testing of BRCA1 and BRCA2 in Asian breast cancer patients identify carriers in those with presumed low risk by family history. Breast Cancer Res Treat 144: 635-642 2014;
  • 19 Malhotra H, Kowtal P, Mehra N. et al. Genetic counseling, testing, and management of HBOC in India: an expert consensus document from Indian Society of Medical and Pediatric Oncology. JCO Glob Oncol 2020; 6: 991-1008
  • 20 Parikh PM, Aggarwal S, Biswas G. et al. Practical clinical consensus guidelines for the management of cancer associated anemia in low- and middle-income countries. South Asian J Cancer 2023; 12 (02) 93-99
  • 21 Phadke SR, Pandey A, Puri RD, Patil SJ. Genetic counseling: the impact in Indian milieu. Indian J Pediatr 2004; 71 (12) 1079-1082
  • 22 National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Genetic/familial high-risk assessment—Breast, Ovarian, and pancreatic (version 1.2020). Accessed July 11, 2024 at: https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf
  • 23 Riley BD, Culver JO, Skrzynia C. et al. Essential elements of genetic cancer risk assessment, counseling, and testing: updated recommendations of the National Society of Genetic Counselors. J Genet Couns 2012; 21 (02) 151-161
  • 24 Allen CG, Roberts M, Guan Y. Exploring predictors of genetic counseling and testing for hereditary breast and ovarian cancer: findings from the 2015 U.S. National Health Interview Survey. J Pers Med 2019; 9 (02) 26
  • 25 Fallowfield L, Jenkins V. Effective communication skills are the key to good cancer care. Eur J Cancer 1999; 35 (11) 1592-1597
  • 26 Robson ME, Bradbury AR, Arun B. et al. American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol 2015; 33 (31) 3660-3667
  • 27 National Pathology Accreditation Advisory Council. Classification of Human Genetic Testing. Canberra, ACT, Australia, Laboratory Accreditation Standards and Guidelines for Nucleic Acid Detection and Analysis; 2007
  • 28 Rantanen E, Hietala M, Kristoffersson U. et al. Regulations and practices of genetic counselling in 38 European countries: the perspective of national representatives. Eur J Hum Genet 2008; 16 (10) 1208-1216
  • 29 Darooei M, Poornima S, Salma BU. et al. Pedigree and BRCA gene analysis in breast cancer patients to identify hereditary breast and ovarian cancer syndrome to prevent morbidity and mortality of disease in Indian population. Tumour Biol 2017; 39 (02) 1010428317694303
  • 30 Rich EC, Burke W, Heaton CJ. et al. Reconsidering the family history in primary care. J Gen Intern Med 2004; 19 (03) 273-280
  • 31 Yip CH, Evans DG, Agarwal G. et al. Global disparities in breast cancer genetics testing, counselling and management. World J Surg 2019; 43 (05) 1264-1270
  • 32 Wood ME, Kadlubek P, Pham TH. et al. Quality of cancer family history and referral for genetic counseling and testing among oncology practices: a pilot test of quality measures as part of the American Society of Clinical Oncology Quality Oncology Practice Initiative. J Clin Oncol 2014; 32 (08) 824-829
  • 33 Brédart A, Kop JL, De Pauw A. et al. Effect on perceived control and psychological distress of genetic knowledge in women with breast cancer receiving a BRCA1/2 test result. Breast 2017; 31: 121-127
  • 34 Mancini J, Noguès C, Adenis C. et al. Impact of an information booklet on satisfaction and decision-making about BRCA genetic testing. Eur J Cancer 2006; 42 (07) 871-881
  • 35 Quinn VF, Meiser B, Kirk J. et al. Streamlined genetic education is effective in preparing women newly diagnosed with breast cancer for decision making about treatment-focused genetic testing: a randomized controlled noninferiority trial. Genet Med 2017; 19 (04) 448-456
  • 36 Randall J, Butow P, Kirk J, Tucker K. Psychological impact of genetic counselling and testing in women previously diagnosed with breast cancer. Intern Med J 2001; 31 (07) 397-405
  • 37 National Cancer Institute. Susceptibility genes. Accessed July 11, 2024 at: https://www.cancer.gov/types/breast/hp/breast-ovarian-genetics-pdq
  • 38 Kowalewski A, Szylberg Ł, Saganek M, Napiontek W, Antosik P, Grzanka D. Emerging strategies in BRCA-positive pancreatic cancer. J Cancer Res Clin Oncol 2018; 144 (08) 1503-1507
  • 39 Singh J, Thota N, Singh S. et al. Screening of over 1000 Indian patients with breast and/or ovarian cancer with a multi-gene panel: prevalence of BRCA1/2 and non-BRCA mutations. Breast Cancer Res Treat 2018; 170 (01) 189-196
  • 40 Agarwal G, Pradeep PV, Aggarwal V, Yip CH, Cheung PS. Spectrum of breast cancer in Asian women. World J Surg 2007; 31 (05) 1031-1040
  • 41 Sharma-Oates A, Shaaban AM, Tomlinson I, Wynne L, Cazier JB, Sundar S. Heterogeneity of germline variants in high risk breast and ovarian cancer susceptibility genes in India. Precis Clin Med 2018; 1 (02) 75-87
  • 42 Thirthagiri E, Lee SY, Kang P. et al. Evaluation of BRCA1 and BRCA2 mutations and risk-prediction models in a typical Asian country (Malaysia) with a relatively low incidence of breast cancer. Breast Cancer Res 2008; 10 (04) R59
  • 43 Gupta S, Rajappa S, Advani S. et al. Prevalence of BRCA1 and BRCA2 mutations among patients with ovarian, primary peritoneal, and fallopian tube cancer in India: a multicenter cross-sectional study. JCO Glob Oncol 2021; 7: 849-861
  • 44 Mittal A, Deo SVS, Gogia A. et al. Profile of pathogenic mutations and evaluation of germline genetic testing criteria in consecutive breast cancer patients treated at a North Indian tertiary care center. Ann Surg Oncol 2022; 29 (02) 1423-1432
  • 45 Pramanik R, Upadhyay A, Khurana S. et al. Comprehensive germline genomic profiling of patients with ovarian cancer: a cross-sectional study. Indian J Med Paediatr Oncol 2022; 43 (04) 361-368
  • 46 Kadri MSN, Patel KM, Bhargava PA. et al. Mutational landscape for Indian hereditary breast and ovarian cancer cohort suggests need for identifying population specific genes and biomarkers for screening. Front Oncol 2021; 10: 568786
  • 47 Chheda P, Pande S, Dama T. et al. Spectrum of germline BRCA mutations in hereditary breast and ovarian cancer syndrome in Indian population: A central reference laboratory experience. Cancer Res Stat Treat 2020; 3: 32-41
  • 48 Mehta A, Vasudevan S, Sharma SK. et al. Germline BRCA1 and BRCA2 deleterious mutations and variants of unknown clinical significance associated with breast/ovarian cancer: a report from North India. Cancer Manag Res 2018; 10: 6505-6516
  • 49 Suryavanshi M, Kumar D, Panigrahi MK, Chowdhary M, Mehta A. Detection of false positive mutations in BRCA gene by next generation sequencing. Fam Cancer 2017; 16 (03) 311-317
  • 50 Stewart MD, Merino Vega D, Arend RC. et al. Homologous recombination deficiency: concepts, definitions, and assays. Oncologist 2022; 27 (03) 167-174
  • 51 Bekos C, Grimm C, Kranawetter M. et al. Reliability of tumor testing compared to germline testing for detecting BRCA1 and BRCA2 mutations in patients with epithelial ovarian cancer. J Pers Med 2021; 11 (07) 593
  • 52 Chandrsekaran D, Sobocan M, Blyuss O. et al. Implementation of multigene germline and parallel somatic genetic testing in epithelial ovarian cancer: SIGNPOST study. Cancers (Basel) 2021; 13: 4344
  • 53 Miller RE, Leary A, Scott CL. et al. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann Oncol 2020; 31 (12) 1606-1622
  • 54 Owens DK, Davidson KW, Krist AH. et al; US Preventive Services Task Force. Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: US Preventive Services Task Force recommendation statement. JAMA 2019; 322 (07) 652-665
  • 55 Vaidyanathan K, Lakhotia S, Ravishankar HM, Tabassum U, Mukherjee G, Somasundaram K. BRCA1 and BRCA2 germline mutation analysis among Indian women from south India: identification of four novel mutations and high-frequency occurrence of 185delAG mutation. J Biosci 2009; 34 (03) 415-422
  • 56 McAlarnen L, Stearns K, Uyar D. Challenges of genomic testing for hereditary breast and ovarian cancers. Appl Clin Genet 2021; 14: 1-9
  • 57 Price KS, Svenson A, King E, Ready K, Lazarin GA. Inherited cancer in the age of next-generation sequencing. Biol Res Nurs 2018; 20 (02) 192-204
  • 58 Ruiz de Garibay G, Gutiérrez-Enríquez S, Garre P. et al. Characterization of four novel BRCA2 large genomic rearrangements in Spanish breast/ovarian cancer families: review of the literature, and reevaluation of the genetic mechanisms involved in their origin. Breast Cancer Res Treat 2012; 133 (01) 273-283
  • 59 Reddy RRS, Ramanujam MV. High throughput sequencing-based approaches for gene expression analysis. Methods Mol Biol 2018; 1783: 299-323
  • 60 Avoiding the common mistakes labs make when launching hereditary cancer panel tests Jeanette McCarthy. Accessed July 11, 2024 at: www.fabricgenomics.com
  • 61 Feliubadaló L, Lopez-Doriga A, Castellsagué E. et al. Next-generation sequencing meets genetic diagnostics: development of a comprehensive workflow for the analysis of BRCA1 and BRCA2 genes. Eur J Hum Genet 2013; 21 (08) 864-870
  • 62 Tucker K. Germline genetic testing: why it matters and where we are failing. Gynaecologic Oncology Consult 2022 . Accessed November 13, 2023 at: https://www.mdedge.com/obgyn/article/258782/gynecologic-cancer/germline-genetic-testing-why-it-matters-and-where-we-are
  • 63 Carvalho CM, Braga LDC, Silva LM, Chami AM, Silva Filho ALD. Germline mutations landscape in a cohort of the state of Minas Gerais, Brazil, in patients who underwent genetic counseling for gynecological and breast cancer. Rev Bras Ginecol Obstet 2023; 45 (02) 74-81
  • 64 Wallace AJ. New challenges for BRCA testing: a view from the diagnostic laboratory. Eur J Hum Genet 2016; 24 (Suppl. 01) S10-S18
  • 65 Jennings LJ, Arcila ME, Corless C. et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn 2017; 19 (03) 341-365
  • 66 Richards S, Aziz N, Bale S. et al; ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 2015; 17 (05) 405-424
  • 67 Roy S, Coldren C, Karunamurthy A. et al. Standards and guidelines for validating next-generation sequencing bioinformatics pipelines: a joint recommendation of the Association for Molecular Pathology and the College of American Pathologists. J Mol Diagn 2018; 20 (01) 4-27
  • 68 Do H, Wong SQ, Li J, Dobrovic A. Reducing sequence artifacts in amplicon-based massively parallel sequencing of formalin-fixed paraffin-embedded DNA by enzymatic depletion of uracil-containing templates. Clin Chem 2013; 59 (09) 1376-1383
  • 69 Hoppe MM, Sundar R, Tan DSP, Jeyasekharan AD. Biomarkers for homologous recombination deficiency in cancer. J Natl Cancer Inst 2018; 110 (07) 704-713
  • 70 Plon SE, Eccles DM, Easton D. et al; IARC Unclassified Genetic Variants Working Group. Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results. Hum Mutat 2008; 29 (11) 1282-1291
  • 71 Meijers-Heijboer H, van Geel B, van Putten WL. et al. Breast cancer after prophylactic bilateral mastectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 2001; 345 (03) 159-164
  • 72 Domchek SM, Friebel TM, Singer CF. et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA 2010; 304 (09) 967-975
  • 73 Kauff ND, Satagopan JM, Robson ME. et al. Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 2002; 346 (21) 1609-1615
  • 74 Zuntini R, Ferrari S, Bonora E. et al. Dealing with BRCA1/2 unclassified variants in a cancer genetics clinic: does cosegregation analysis help?. Front Genet 2018; 9: 378
  • 75 Neff RT, Senter L, Salani R. BRCA mutation in ovarian cancer: testing, implications and treatment considerations. Ther Adv Med Oncol 2017; 9 (08) 519-531
  • 76 Nagahashi M, Shimada Y, Ichikawa H. et al. Formalin-fixed paraffin-embedded sample conditions for deep next generation sequencing. J Surg Res 2017; 220: 125-132
  • 77 Cazzato G, Caporusso C, Arezzo F. et al. Formalin-fixed and paraffin-embedded samples for next generation sequencing: problems and solutions. Genes (Basel) 2021; 12 (10) 1472
  • 78 Moore DA, Balbi K, Ingham A, Arkenau HT, Bennett P. Analysis of a large cohort of non-small cell lung cancers submitted for somatic variant analysis demonstrates that targeted next-generation sequencing is fit for purpose as a molecular diagnostic assay in routine practice. J Clin Pathol 2018; 71 (11) 1001-1006
  • 79 Rajkumar T, Meenakumari B, Mani S, Sridevi V, Sundersingh S. Targeted resequencing of 30 genes improves the detection of deleterious mutations in South Indian women with breast and/or ovarian cancers. Asian Pac J Cancer Prev 2015; 16 (13) 5211-5217
  • 80 Borg A, Haile RW, Malone KE. et al. Characterization of BRCA1 and BRCA2 deleterious mutations and variants of unknown clinical significance in unilateral and bilateral breast cancer: the WECARE study. Hum Mutat 2010; 31 (03) E1200-E1240
  • 81 Exchange BRCA. . Summary view. Accessed July 11, 2024 at: https://brcaexchange.org/
  • 82 Lindor NM, Goldgar DE, Tavtigian SV, Plon SE, Couch FJ. BRCA1/2 sequence variants of uncertain significance: a primer for providers to assist in discussions and in medical management. Oncologist 2013; 18 (05) 518-524
  • 83 Ledermann J, Harter P, Gourley C. Correction to Lancet Oncol 2014; 15: 856. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 2015; 16 (04) e158
  • 84 Hennessy BT, Timms KM, Carey MS. et al. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol 2010; 28 (22) 3570-3576
  • 85 Ledermann J, Harter P, Gourley C. et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 2014; 15 (08) 852-861
  • 86 Sandhu SK, Schelman WR, Wilding G. et al. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol 2013; 14 (09) 882-892
  • 87 Agarwal A, Baghmar S, Dodagoudar C. et al. PARP inhibitor in platinum-resistant ovarian cancer: single-center real-world experience. JCO Glob Oncol 2021; 7: 506-511
  • 88 Vergote I, Ray-Coquard I, Anderson DM. et al. Population-adjusted indirect treatment comparison of the SOLO1 and PAOLA-1/ENGOT-ov25 trials evaluating maintenance olaparib or bevacizumab or the combination of both in newly diagnosed, advanced BRCA-mutated ovarian cancer. Eur J Cancer 2021; 157: 415-423
  • 89 Kanjanapan Y, Lheureux S, Oza AM. Niraparib for the treatment of ovarian cancer. Expert Opin Pharmacother 2017; 18 (06) 631-640
  • 90 Vendrell JA, Ban IO, Solassol I. et al. Differential sensitivity of germline and somatic BRCA variants to PARP inhibitor in high-grade serous ovarian cancer. Int J Mol Sci 2023; 24 (18) 14181
  • 91 Terraf P, Pareja F, Brown DN. et al. Comprehensive assessment of germline pathogenic variant detection in tumor-only sequencing. Ann Oncol 2022; 33 (04) 426-433
  • 92 Tew WP, Lacchetti C, Ellis A. et al. PARP inhibitors in the management of ovarian cancer: ASCO guideline. J Clin Oncol 2020; 38 (30) 3468-3493
  • 93 Rebbeck TR, Lynch HT, Neuhausen SL. et al; Prevention and Observation of Surgical End Points Study Group. Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 2002; 346 (21) 1616-1622
  • 94 Carbine NE, Lostumbo L, Wallace J, Ko H. Risk-reducing mastectomy for the prevention of primary breast cancer. Cochrane Database Syst Rev 2018; 4 (04) CD002748
  • 95 Lostumbo L, Carbine NE, Wallace J. Prophylactic mastectomy for the prevention of breast cancer. Cochrane Database Syst Rev 2010; CD002748 (11) CD002748
  • 96 Heemskerk-Gerritsen BA, Seynaeve C, van Asperen CJ. et al; Hereditary Breast and Ovarian Cancer Research Group Netherlands. Breast cancer risk after salpingo-oophorectomy in healthy BRCA1/2 mutation carriers: revisiting the evidence for risk reduction. J Natl Cancer Inst 2015; 107 (05) djv033
  • 97 Kotsopoulos J, Huzarski T, Gronwald J. et al; Hereditary Breast Cancer Clinical Study Group. Bilateral oophorectomy and breast cancer risk in BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 2016; 109 (01) djw177
  • 98 Terry MB, Daly MB, Phillips KA. et al. Risk-reducing oophorectomy and breast cancer risk across the spectrum of familial risk. J Natl Cancer Inst 2019; 111 (03) 331-334
  • 99 NCCN. Accessed July 11, 2024 at: https://www.nccn.org/guidelines/guidelines-detail?category=2&id=1420
  • 100 Saslow D, Boetes C, Burke W. et al; American Cancer Society Breast Cancer Advisory Group. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin 2007; 57 (02) 75-89
  • 101 King MC, Wieand S, Hale K. et al; National Surgical Adjuvant Breast and Bowel Project. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA 2001; 286 (18) 2251-2256
  • 102 Tung NM, Boughey JC, Pierce LJ. et al. Management of hereditary breast cancer: American Society of Clinical Oncology, American Society for Radiation Oncology, and Society of Surgical Oncology Guideline. J Clin Oncol 2020; 38 (18) 2080-2106
  • 103 Pesce C, Liederbach E, Wang C, Lapin B, Winchester DJ, Yao K. Contralateral prophylactic mastectomy provides no survival benefit in young women with estrogen receptor-negative breast cancer. Ann Surg Oncol 2014; 21 (10) 3231-3239
  • 104 Wong SM, Freedman RA, Sagara Y, Aydogan F, Barry WT, Golshan M. Growing use of contralateral prophylactic mastectomy despite no improvement inlong term survival for invasive breast cancer. Ann Surg 2017; 265 (03) 581-589
  • 105 Son KA, Lee DY, Choi D. Association of BRCA mutations and anti-müllerian hormone level in young breast cancer patients. Front Endocrinol (Lausanne) 2019; 10: 235
  • 106 Rădoi VE, Țurcan M, Maioru OV. et al. Homologous recombination deficiency score determined by genomic instability in a Romanian cohort. Diagnostics (Basel) 2023; 13 (11) 1896
  • 107 Robson ME, Tung N, Conte P. et al. OlympiAD final overall survival and tolerability results: Olaparib versus chemotherapy treatment of physician's choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol 2019; 30 (04) 558-566
  • 108 Litton JK, Hurvitz SA, Mina LA. et al. Talazoparib versus chemotherapy in patients with germline BRCA1/2-mutated HER2-negative advanced breast cancer: final overall survival results from the EMBRACA trial. Ann Oncol 2020; 31 (11) 1526-1535
  • 109 von Minckwitz G, Schneeweiss A, Loibl S. et al. Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): a randomised phase 2 trial. Lancet Oncol 2014; 15 (07) 747-756
  • 110 Sikov WM, Berry DA, Perou CM. et al. Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance). J Clin Oncol 2015; 33 (01) 13-21
  • 111 Tutt ANJ, Garber JE, Kaufman B. et al; OlympiA Clinical Trial Steering Committee and Investigators. Adjuvant Olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med 2021; 384 (25) 2394-2405
  • 112 Aschenbrenner DS. New adjuvant treatment for high-risk early breast cancer. Am J Nurs 2022; 122 (07) 26
  • 113 DiSilvestro P, Banerjee S, Colombo N. et al; SOLO1 Investigators. Overall survival with maintenance olaparib at a 7-year follow-up in patients with newly diagnosed advanced ovarian cancer and a BRCA mutation: the SOLO1/GOG 3004 trial. J Clin Oncol 2023; 41 (03) 609-617
  • 114 Pujade-Lauraine E, Ledermann JA, Selle F. et al; SOLO2/ENGOT-Ov21 investigators. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18 (09) 1274-1284
  • 115 Fabbro M, Moore KN, Dørum A. et al. Efficacy and safety of niraparib as maintenance treatment in older patients (≥ 70 years) with recurrent ovarian cancer: Results from the ENGOT-OV16/NOVA trial. Gynecol Oncol 2019; 152 (03) 560-567
  • 116 Oza AM, Tinker AV, Oaknin A. et al. Antitumor activity and safety of the PARP inhibitor rucaparib in patients with high-grade ovarian carcinoma and a germline or somatic BRCA1 or BRCA2 mutation: Integrated analysis of data from Study 10 and ARIEL2. Gynecol Oncol 2017; 147 (02) 267-275
  • 117 Moore K, Colombo N, Scambia G. et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 2018; 379 (26) 2495-2505
  • 118 Valarmathi MT, A A, Deo SS, Shukla NK, Das SN. BRCA1 germline mutations in Indian familial breast cancer. Hum Mutat 2003; 21 (01) 98-99
  • 119 Saxena S, Chakraborty A, Kaushal M. et al. Contribution of germline BRCA1 and BRCA2 sequence alterations to breast cancer in Northern India. BMC Med Genet 2006; 7: 75
  • 120 Syamala V, Sreeja L, Syamala VS. et al. Novel germline mutations in BRCA2 gene among 96 hereditary breast and breast-ovarian cancer families from Kerala, South India. J Cancer Res Clin Oncol 2007; 133 (11) 867-874
  • 121 Soumittra N, Meenakumari B, Parija T. et al. Molecular genetics analysis of hereditary breast and ovarian cancer patients in India. Hered Cancer Clin Pract 2009; 7 (01) 13

Zoom Image
Fig. 1 Representative pedigree chart of HBOC family. HBOC, hereditary breast ovarian cancer.