Skip to main content

REVIEW article

Front. Reprod. Health, 15 December 2023
Sec. Assisted Reproduction
This article is part of the Research Topic The Evolution of PGT-M in Diagnostics and Therapeutics View all 4 articles

Genetic counseling for pre-implantation genetic testing of monogenic disorders (PGT-M)

\r\nFiruza Parikh
&#x;Firuza Parikh1*Arundhati Athalye,&#x;Arundhati Athalye1,†Prochi Madon,&#x;Prochi Madon1,‡Meenal Khandeparkar,
Meenal Khandeparkar1,§Dattatray Naik,
Dattatray Naik1,§Rupesh Sanap,
Rupesh Sanap1,§Anuradha Udumudi,&#x;\r\nAnuradha Udumudi2,‡
  • 1Department of Assisted Reproduction and Genetics, Jaslok-FertilTree International Fertility Centre, Jaslok Hospital and Research Centre, Mumbai, India
  • 2GeneTech Laboratory, Banjara Hills, Hyderabad, India

Pre-implantation genetic testing (PGT) is a vital tool in preventing chromosomal aneuploidies and other genetic disorders including those that are monogenic in origin. It is performed on embryos created by intracytoplasmic sperm injection (ICSI). Genetic counseling in the area of assisted reproductive technology (ART) has also evolved along with PGT and is considered an essential and integral part of Reproductive Medicine. While PGT has the potential to prevent future progeny from being affected by genetic conditions, genetic counseling helps couples understand and adapt to the medical, psychological, familial and social implications of the genetic contribution to disease. Genetic counseling is particularly helpful for couples with recurrent miscarriages, advanced maternal age, a partner with a chromosome translocation or inversion, those in a consanguineous marriage, and those using donor gametes. Partners with a family history of genetic conditions including hereditary cancer, late onset neurological diseases and with a carrier status for monogenic disorders can benefit from genetic counseling when undergoing PGT for monogenic disorders (PGT-M). Genetic counseling for PGT is useful in cases of Mendelian disorders, autosomal dominant and recessive conditions and sex chromosome linked disorders and for the purposes of utilizing HLA matching technology for creating a savior sibling. It also helps in understanding the importance of PGT in cases of variants of uncertain significance (VUS) and variable penetrance. The possibilities and limitations are discussed in detail during the sessions of genetic counseling.

Introduction

Pre-implantation genetic testing (PGT) is vital in preventing chromosomal aneuploidies and other genetic disorders including rare genetic monogenic conditions in human embryos created by intracytoplasmic sperm injection (ICSI). The utility of PGT in infertility management has steadily gained importance over the last two decades. Although the practice of genetic counseling originated in the 1960s along with the emergence of prenatal testing, genetic counseling in the area of assisted reproductive technology (ART) has evolved as an integral part of the PGT process and is widely used in reproductive medicine (1).

Genetic counseling is defined by the National Society of Genetic Counselors, USA, as a process of helping people understand and adapt to the medical, psychological and familial implications of the genetic contributions to disease. This process integrates the following: (a) Interpretation of the family and medical history to assess the chance of disease occurrence or recurrence. (b) Education about inheritance, testing, management, prevention, available resources and current research. (c) Counseling to facilitate informed choices and adaptation to the risk or condition (2, 3). Genetic counseling is specially helpful for families facing complex situations that may arise during the process of PGT for Monogenic disorders (PGT-M). The current review is an attempt to introduce readers to some situations that can arise while offering PGT-M.

While the advancement of diagnostic technologies offers prevention of genetic disorders and birth defects, the dissemination and interpretation of information on the disease-causing variations and their solutions can best be served with genetic counseling. With PGT emerging as an early alternative to post-conception prenatal diagnosis, couples are in a better position to plan a pregnancy with the help of in vitro fertilization (IVF) driven PGT. Preimplantation genetic technology aims to prevent the trauma of repeated termination of pregnancies in cases where genetic abnormalities in the fetus are diagnosed much later by prenatal testing. Genetic counseling plays an important role in communicating the benefits, limitations and complexities of PGT in an IVF set up in a non-directive way, with empathy.

Counseling during the PGT procedure

PGT is an integral part of an experienced IVF centre and is done in collaboration with a genetics laboratory with skills to accurately report on the results of testing 1–8 embryonic cells. On day 5–6 after the ICSI procedure, about 5–8 herniated trophectoderm cells are biopsied from a site on the blastocyst away from the inner cell mass by an experienced embryologist. The biopsied cells from each embryo are transferred into separate tubes and sent to specialized genetic laboratories for testing (4, 5). All concerns of patients related to the invasive nature of micromanipulation and lack of long-term safety outcome data should be addressed with transparency. Reassurance about the safety of recent biopsy techniques of the trophectoderm compared to previously used techniques should be shared (6). After biopsy, the embryos are vitrified until the availability of results. Although the risks associated with thawing of vitrified embryos is minimal, couples should be made aware of them (7).

PGT which was previously called pre-implantation genetic diagnosis (PGD) includes three terms, PGT-A, PGT-SR and PGT-M (810). PGT screens embryos for chromosomal aneuploidies (PGT-A) thereby increasing the chances of implantation and reducing pregnancy loss (11). PGT for structural rearrangement (PGT-SR) including reciprocal balanced translocations, inversions or insertions is helpful for individuals with chromosomal rearrangements. PGT for monogenic diseases (PGT-M) has the potential to offer reproductive options to couples who are at an increased risk of having progeny with single gene disorders such as thalassemia, sickle cell anemia, cystic fibrosis and Huntington disease to name a few. PGT-A is also carried out together with PGT-M to increase the chances of success, by selecting euploid unaffected embryos for transfer. This review is limited to the discussion of PGT-M.

Pre-implantation genetic testing for monogenic disorders (PGT-M)

In 1990, Handyside's group first reported live births using IVF followed by PGD for an X-linked disorder (12). After 2005, different methodologies evolved resulting in a plethora of techniques capable of diagnosing genetic disorders. These were quickly adopted in PGT. They include haplotyping (13), comparative genomic hybridization (aCGH) (14), next generation sequencing (NGS) (15), karyomapping (16) and linkage analysis (17). PGT-M is presently available for identifying pathogenic or likely pathogenic disease causing variants in embryos for all known monogenic disorders (18). These disorders encompass both common Mendelian conditions such as beta thalassemia, cystic fibrosis, hemophilia, sickle cell anemia, Huntington's chorea, Duchenne muscular dystrophy, as well as rarer genetic diseases like methylmalonic acidemia and lysosomal storage disorders. Additionally, specific mitochondrial disorders like Leigh syndrome where nuclear DNA is involved (19, 20), are also included. The aim of genetic counseling is to effectively convey the advantages and constraints of PGT while ensuring that patients have comprehensive information to make an informed decision, taking into account the outcomes derived from the test. Some of the important features of PGT-M genetic counseling are discussed in this review.

Pre-PGT-M genetic counseling

It is important for all couples considering PGT-M to participate in genetic counseling and share their family history, medical records and results of molecular studies carried out for family members. A prerequisite for PGT-M testing is that the disease-causing variant is identified and classified as per American College of Medical Genetics (ACMG) guidelines for pathogenicity before initiating PGT-M (2123). Based on the pedigree analysis and review of reports, the genetic counselor is in a position to discuss the inheritance pattern of the condition in the family, risks to the offspring and available preventive options including prenatal or pre-implantation genetic testing. The benefits of PGT vs. its risks, limitations, and cost of testing should be communicated to the family. Informed written consent is necessary and should be taken prior to initiating the PGT procedure.

Determination of the disease causing variation and evaluation of the inheritance pattern are prerequisites for PGT-M

Most of the Mendelian disorders are caused by disease causing variations in a single gene (monogenic). The inheritance of these conditions can be autosomal dominant, autosomal recessive, X-linked dominant or X-linked recessive. The involvement of the genetic counselor begins by looking at the phenotype of the affected individual in the family, doing a dysmorphology assessment, reviewing the medical records, determining the inheritance pattern and ordering tests to identify the disease-causing variation along with its inheritance. Based on the phenotype analysis, the counselor follows a genetic diagnostic workflow which is economical and best suited for the case (Figures 15). The following case examples demonstrate the adopted workflow in frequent counseling scenarios:

Figure 1
www.frontiersin.org

Figure 1. Genetic diagnostic workflow for PGT-M of common known variants in certain disorders.

Figure 2
www.frontiersin.org

Figure 2. Genetic diagnostic workflow for PGT-M of autosomal dominant conditions.

Figure 3
www.frontiersin.org

Figure 3. Genetic diagnostic workflow for PGT-M of autosomal recessive conditions.

Figure 4
www.frontiersin.org

Figure 4. Genetic diagnostic workflow for PGT-M of X-linked recessive conditions.

Figure 5
www.frontiersin.org

Figure 5. Genetic diagnostic workflow for PGT-M of X-linked dominant conditions.

(a) Common genetic variations such as ΔF508 in cystic fibrosis [c.1521_1523del (p.Phe508del)] and c.20A>T (p.Glu6Val) in sickle cell anemia can be checked by simple molecular techniques such as polymerase chain reaction (PCR) (2427). If the variation is confirmed either in the index case or the carrier parents, PGT-M can be offered for the condition (Figure 1).

(b) If common genetic variations are absent in a case with clear cystic fibrosis or sickle cell anemia phenotypes, the genetic counselor will order tests with wider scope such as full gene sequencing of CFTR or beta globin gene to identify rare pathogenic variations (28, 29). Many times it is seen that the individual with sickle cell anemia may actually have sickle cell thalassemia with a sickle cell heterozygous variation and a thalassemia heterozygous variation (30). Full gene sequencing will help to identify such cases. Based on these results, the option of PGT-M can be discussed with the family (Figure 1).

(c) In autosomal dominant conditions such as history of a child with achondroplasia, confirmed by a genetic test, the recurrence risk is usually low as parents are unaffected. The sporadic occurrence of achondroplasia and low recurrence risk is explained to the couple and PGT-M may not be offered. The extreme rare condition of gonadal mosaicism, where parents may be unaffected carriers but produce gametes with a variation due to the presence of a variation only in gonadal tissue should be discussed. In such condition PGT-M can be offered after counseling. However, in a homozygous state, achondroplasia is usually embryonically lethal (31) (Figure 2).

(d) If a parent has an early onset autosomal dominant Alzheimer, Huntington disease or Marfan syndrome, the risk of recurrence for offspring is 50% hence the PGT-M option is explained to the couple (3234) (Figure 2).

(e) For autosomal recessive disorders, the workflow will include identification of a recessive variation in the index child or identification of the carrier status in both partners (35, 36). Examples of recessive disorders include thalassemia, cystic fibrosis and albinism (37). A recurrence risk of 25% for every pregnancy is discussed with the family during the counseling session based on the confirmation of a recessive variation in the index case or carrier status of both partners by genetic tests. In some instances where the proband or affected family member is unavailable, carrier screening is recommended for parents to identify the general risk of inheriting recessive disorders that are not indicated by family history. Carrier screening can be offered for selective targeted disorders by targeted NGS panels or for all recessive disorders using whole exome sequencing (WES) methodologies. This testing is particularly helpful in consanguineous marriages. Genetic counseling in the presence of consanguinity is vital in order to explain the possibility of more than one recessive disorder risk in the offspring because of the higher proportion of shared genetic material between the partners (38) (Figure 3).

(f) In cases such as Duchene muscular dystrophy (DMD) or spinal muscular atrophy (SMA), the multiplex ligation dependent probe amplification (MLPA) technology is utilized to identify exon level deletions or duplications. Confirmation of pathogenic homozygous deletion/duplication in affected individuals or hemizygous/heterozygous deletion/duplication in carrier parents will open up the discussion on the option of PGT-M (Figure 4) (39, 40).

(g) For X-linked recessive conditions like hemophilia, where males are mainly affected, the counselor will recommend identification of the gene variant in the affected male child or in the female partner, who is usually an unaffected carrier with a variation on one of her X chromosomes, the other X chromosome being normal. A 50% risk to the male offspring is explained as the risk that she passes on when she transmits an X chromosome with the variant. Female offspring are usually unaffected like their mother (41) (Figure 4).

(h) In certain scenarios the counseling session will clarify that there is no risk to the offspring and PGT-M may not be required. For instance, if the male partner's brother has DMD which is an X-linked recessive condition, there is no risk for the couple as the male partner has a normal phenotype being devoid of the variation on his X chromosome (Figure 4).

(i) Although rare, X-linked dominant conditions such as, Incontinentia pigmenti and Rett syndrome equally affect males and females. Males with X-linked dominant condition are affected more severely with low survivability than females. In many cases in-utero demise is a common phenomenon observed in affected male fetuses. An affected mother will have a 50% chance of passing on the dominant gene variant to either male or female offsping. In case of an affected father there is 50% risk for female offspring and no risk for male offspring as the father does not contribute the X-chromosome to a male child (Figure 5).

(j) There are many other complex disorders with multi-gene etiology and for their diagnosis including differential diagnosis, NGS based WES is used. The test relies on sequencing of genomic exons or the coding regions of the genome and is usually recommended for the affected family member (Index or proband) (42, 43). Couple carrier screening can also be offered. Based on the test results, the genetic counselor would guide the family and patients for follow-up tests which could be either in the prenatal or pre-implantation period.

(k) There are many disorders for which PGT-M was successfully offered (4446).

The dilemma of variants of uncertain significance (VUS)—pathogenic or benign?

With the advancement of NGS technologies allowing rapid and inexpensive sequencing of the human genome, we are witnessing its rapid adoption in clinics for couples as a preliminary genetic work up by choice, before starting any fertility treatment. A change in the genetic code is usually called a variant and thousands of variants are identified in the process of WES or whole genome sequencing. These variants are classified into five tiers: pathogenic (P), likely pathogenic (LP), variant of uncertain significance (VUS), likely benign (LB), and benign (B) as per ACMG guidelines. Pathogenic and likely pathogenic variants are those that are strongly associated with disease etiology hence offering PGT-M in such situations is justified. Benign and likely benign are those variants that may not be disease causing. Therefore, PGT-M is not offered. Variants that are not classified either as pathogenic or benign are reported as VUS (21). A VUS is a genetic change whose clinical impact is not yet well understood, either because its effect on the gene's function is not fully known or because there is lack of sufficient data for its definitive classification as benign or pathogenic due to a low number of cases reported or due to conflicting results in literature. While many laboratory policies reject PGT-M requests for VUS, some labs accept them, provided the patients clearly understand the implications of testing the VUS (47). An in-depth counseling session will help couples understand these details and a clear documentation of the same communication between the genetic counselor and patients should be made available to the lab along with the signed consent form. It should be noted that the classification of a variant may change over time to likely pathogenic or likely benign with increasing availability of data on the gene variant. Laboratories providing sequencing and reporting services may contact families if there is a change in classification of the variant and issue a revised report. Patients should be encouraged to attend a genetic counseling session with a revised report. Sometimes testing extended family members can help in resolving the issue of pathogenicity of the VUS. This is done by determining whether variants are shared by other affected or unaffected individuals. However, this requires a prolonged work-up of multiple family members and may not be practical in all cases. Considering the above issues with VUS, the focus of the genetic counselor should be to address the uncertainty in such a way that the couple understands the implications in order to make an informed decision.

The process of pre-PGT-M workup

PGT-M optimization includes designing laboratory primers and pre-work-up using DNA of extended family members. The laboratory usually carries out a linkage-based analysis using multiple genetic markers on parents and available family members before testing the embryos. In some cases, the genetic counselor may involve more team members including laboratory personnel and clinical experts in the discussion in order to determine the feasibility of testing the embryos (48). PGT-M may sometimes be complex and difficult in cases of de novo variants, unconfirmed VUS in extended family members, unavailability of relatives, refusal for testing by relatives, in the presence of complex gene loci and other technical difficulties.

Genetic counseling for mitochondrial disorders

Mitochondrial disorders mostly result in metabolic problems. They involve multiple organs with variable disease onset and severity. Variations in both nuclear DNA and mitochondrial DNA (mtDNA) are responsible for mitochondrial disorders.

Disorders caused by variations in nuclear DNA usually follow an autosomal recessive pattern of inheritance, although rarely an autosomal dominant pattern is seen. Prenatal diagnosis and PGT are available to prevent the birth of another severely affected child in such cases of nuclear gene associated mitochondrial disorders (Variation in SURF1 gene for Leigh Syndrome) (49). For de novo variations, the recurrence risk is low and couples can be counseled accordingly.

However in 15%–25% of cases, mitochondrial diseases are caused by mtDNA variations, which can be either de novo or maternally inherited. Mitochondrial DNA sequencing is used to confirm the variations. But for maternally inherited mtDNA variations, the recurrence risk is often unpredictable because of random presence of heteroplasmy (a mixture of normal and variation copies in mtDNA) or homoplasmy (all copies of the mitochondrial genome are identical with or without the variation) in cells. In addition, mtDNA variations are associated with a variable phenotype which leads to difficulty in predicting the phenotypic outcome. The option of oocyte donation may be discussed to prevent transmission of the disorder to the offspring. Discussion of complex mitochondrial inheritance needs extended sessions during genetic counseling (50). Recently, mitochondrial replacement therapy (MRT) has become available for clinical application as an alternative to prevent the transmission of heteroplasmic and homoplasmic mtDNA variations (51).

Genetic counseling for inherited cancer predisposition syndromes

Individuals are considered to be candidates for inherited cancer predisposition syndrome risk assessment if they have a personal and/or a family history on the maternal or paternal side or if they have clinical characteristics with features suggestive of inherited cancer predisposition syndrome. Some examples are familial breast and ovarian cancer, Lynch syndrome, Peutz–Jeghers syndrome and inherited retinoblastoma. Testing the affected individual or an individual at risk is essential to determine the inheritance pattern and identify the risk for future generations. Although prenatal and pre implantation testing can be offered to families at a high risk, it must be kept in mind that testing is done for germline cancer susceptible variants that increase the risk for malignancy and not for somatic variations. PGT can limit the transmission of the inherited cancer gene variation to future generations (5255). It is essential to discuss the challenges that may be encountered including the age of onset, variable penetrance and phenotype variability. Both pre-test and post-test counseling sessions are recommended, so that the individuals are made aware of the details of the genetic report's cancer predisposition for appropriate planning of PGT-M.

Genetic counseling for adult-onset genetic disease

PGT for late adult-onset diseases is complex with a wide range of ethical, legal, social, and policy issues. PGT-M for late adult-onset conditions is a controversial subject with debatable arguments on both sides. The Ethics Committee of the American Society for Reproductive Medicine (ASRM) states that PGT-M for adult-onset conditions is ethically justified when the condition is serious, and no safe, effective interventions are available. The committee however cautions about technological issues, complexity of the scientific, psychological and social aspects and emphasizes on the strong role of an experienced genetic counselor in the process (56). Examples of adult-onset genetic conditions are Huntington disease, myotonic dystrophy, spinocerebellar ataxia, Charcot-Marie-Tooth disease, adult-onset metachromatic leukodystrophy, neurofibromatosis and hereditary cancer predisposition syndromes. While patients with a positive family history seek to understand their carrier status for late onset diseases, sometimes the associated variants are identified in WES tests as secondary or incidental findings. Labs disclose such information only to individuals who consent to know their risk. Once the risk is identified, the adult may choose not to pass on the variant to the offspring and might seek PGT-M for the condition. Many labs accept such a request with the individual's consent. However, one important aspect to consider when discussing PGT-M for late adult-onset genetic diseases is the issue of penetrance. If some individuals with the variant do not develop features of the disorder while others do, the condition is said to have reduced or incomplete penetrance. In such cases, ethical and policy considerations including reproductive liberties take a central place in the discussion. If the variation is identified in an affected grandparent, the children in the reproductive age group could get pre-symptomatic testing done with a view to avoid passing on the deleterious gene to the next generation. Occasionally, these couples may not want to know the result of the pre-symptomatic test, but may want to do PGT anyway, in order to avoid transmission of the pathogenic variant to their children. This is feasible and can be done after discussing non-disclosure protocols and taking appropriate informed consent. The genetic counselors must consider all such situations and provide information to patients in order to facilitate non-directive decision making (57, 58).

Genetic counseling for polygenic disorders (PGT-P)

Polygenic diseases are complex diseases that are influenced by the combined effects of many genes along with an environmental contribution. Examples include commonly occurring diseases, such as coronary heart disease and type 2 diabetes, inherited cancer predisposition syndrome and schizophrenia. Unlike PGT-M, PGT for polygenic disease (PGT-P) represents a further level of complexity in which multiple genes are tested and an associated polygenic risk score (PRS) is established using genome-wide association studies (GWAS). PRS estimates the genetic risk of an individual for a disease or trait, calculated by aggregating the effect of many common variants associated with the condition. Assigning a PRS to an embryo is possible because of available large datasets of people with each disease. Clinical implementation of PRS may be useful in cohorts where there is a higher prior probability of disease, for example, in early stages of diseases to assist in the diagnosis or to inform individuals regarding treatment choices (56, 5962). The pros and cons of PGT-P are highly debated among the scientific circles and there are vocal proponents and opponents on both sides arguing cost to benefit justification (63). Although a few companies are already offering such services, genetic counselors should caution families of the limitations such as uncertainty of the risks, non-availability of data for many population groups, absence of long term studies and the fact that it is only predictive with a risk score and not diagnostic. Currently these limitations lead to attenuated feasibility of prenatal or PGT-P (64).

Genetic counseling for epigenetic disorders

The epigenome surrounds the genome or the genetic component of the cell and influences the mechanism of turning on and off the genes by condensation, folding and unfolding of DNA. Histones, methyl groups and microRNAs constitute the epigenome. Many human diseases including neurodevelopment conditions, cancer and life style disorders are influenced by epigenetic changes which are dependent on the environment. Although the search for new fetal epigenetic markers and the clinical implementation of epigenetic approaches for noninvasive prenatal diagnosis are underway (65), prenatal and PGT options are not currently feasible. Pre-pregnancy parental lifestyle, post pregnancy maternal care and early childhood nurturing, make for an optimal epigenome that can protect individuals throughout their life span (66).

Post PGT-M genetic counseling

A post-test genetic counseling session explaining the PGT-M results and their interpretation should be carried out. Usually, along with testing for the monogenic disorders, PGT-A is also offered on the embryos so that the risk of aneuploidy is also ascertained (48). Aneuploid embryo with a normal or unaffected carrier status for the tested genetic condition is chosen for transfer to the uterus at a time when the endometrium is most receptive. Other normal embryos are cryopreserved for future use. It is important to counsel individuals about the rare false negative results, however rare, caused by both technical limitations and complex biological processes. For example, a disintegrated DNA sample might fail to amplify both the alleles of the variant simultaneously, resulting in a false homozygous report. This phenomenon of amplification of only one allele, instead of both, is called as allele drop-out. In vivo, the early embryonic divisions are prone to errors and may give rise to sporadic genetic variants that may be pathogenic in the fetus. Hence diagnostic prenatal tests by chorionic villus sampling (CVS) or amniocentesis processes should be communicated during counseling in a simple language with emphasis on the need of post-conception testing. The entire process of genetic counseling in PGT-M is summarized in a flow chart in Figure 6. It gives a broad outline of steps usually followed by the genetic counselor before initiating the tests and after evaluating the type of result (pathogenic, likely pathogenic, uncertain significance variants), late onset disease diagnosis and secondary findings.

Figure 6
www.frontiersin.org

Figure 6. Summary of end to end genetic counseling work flow for PGT-M.

Genetic counseling for PGT-M for a savior sibling

A savior sibling is usually considered by parents when a child needs a bone marrow transplant and finding a compatible donor is a challenge. The savior baby is generally created through IVF by testing the embryos by human leukocyte antigen (HLA) typing and using the embryos that are 100% HLA matched with the older affected sibling. A wide range of genetic disorders can be treated by savior siblings including blood disorders like thalassemia, leukemia, bone marrow disorders, immune deficiencies and certain cancers. It is important to select an embryo which is not only 100% HLA match but is also devoid of the pathogenic disease variation present in the older sibling. Haematopoietic stem cells collected from the umbilical cord blood or the bone marrow of the HLA-matched donor sibling born are used for transplantation to cure the affected sibling (67). HLA typing of ART-created embryos was first reported in 2001 (68) and with several encouraging results reported worldwide, savior sibling creation is now offered in several IVF clinics (69). The genetic counselor will have to discuss ethical, technical and financial implications of a savior sibling during the genetic counseling session (70).

Conclusion

The role of genetic counseling for couples undergoing PGT-M is crucial and must be offered by trained, qualified genetic counselors. Usually the information gathered during a genetic counseling session helps the counselor to not only offer PGT-M option based on the case details, but also to select appropriate technology or workflow for pre-PGT-M work up. Genetic counselors will have to consider the inheritance pattern, availability, or non-availability of index case, family history and clinical parameters before giving out the recommendations. Complex genetic disorders including mitochondrial diseases and polygenic conditions must be addressed with care. Communicating genetic test results to the family and helping them understand the implications is an important part of the sessions. The genetic counselor plays a pivotal role in establishing a close collaboration between genetic labs and ART centers which is paramount for successful implementation of PGT-M. As couples would be going through complex IVF treatment together with genetic testing, it is expected that they may be sensitive and emotionally charged and may ask abundant questions. A genetic counselor should be proficient with the language of the patient/family, have empathy, should be willing to listen and explain, be supportive, non-directive, and use effective communication through charts and videos. Genetic counselors should align their discussion with ethical, religious issues and the rules and regulations of the governing body of the residing country.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of interest

AU is employed by GeneTech Laboratory, Hyderabad, India.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Parikh FR, Athalye AS, Kulkarni DK, Sanap RR, Dhumal SB, Warang DJ, et al. Evolution and utility of preimplantation genetic testing for monogenic disorders in assisted reproduction—a narrative review. J Hum Reprod Sci. (2021) 14:329–39. doi: 10.4103/jhrs.jhrs_148_21

PubMed Abstract | Crossref Full Text | Google Scholar

2. Epstein CJ, Childs B, Fraser FC, McKusick VA, Miller JR, Motulsky AG, et al. Genetic counseling. Am J Hum Genet. (1975) 27:240–2. PMCID: PMC1762759; PMID: 1124768

PubMed Abstract | Google Scholar

3. Resta R, Biesecker BB, Bennett RL, Blum S, Hahn SE, Strecker MN, et al. A new definition of genetic counseling: national society of genetic counselors’ task force report. J Genet Couns. (2006) 15:77–83. doi: 10.1007/s10897-005-9014-3

PubMed Abstract | Crossref Full Text | Google Scholar

4. Monk M, Muggleton-Harris AL, Rawlings E, Whittingham DG. Pre-implantation diagnosis of HPRT-deficient male and carrier female mouse embryos by trophectoderm biopsy. Hum Reprod. (1988) 3:377–81. doi: 10.1093/oxfordjournals.humrep.a136711

PubMed Abstract | Crossref Full Text | Google Scholar

5. Scott RT Jr, Upham KM, Forman EJ, Zhao T, Treff NR. Cleavage-stage biopsy significantly impairs human embryonic implantation potential while blastocyst biopsy does not: a randomized and paired clinical trial. Fertil Steril. (2013) 100:624–30. doi: 10.1016/j.fertnstert.2013.04.039

PubMed Abstract | Crossref Full Text | Google Scholar

6. Scott KL, Hong KH, Scott RT Jr. Selecting the optimal time to perform biopsy for preimplantation genetic testing. Fertil Steril. (2013) 100(3):608e14. doi: 10.1016/j.fertnstert.2013.07.004

Crossref Full Text | Google Scholar

7. Mounts EL, Besser AG. Genetic counseling for preimplantation genetic testing (PGT): practical and ethical challenges. In: Palermo G, Sils S, editors. Human embryos and preimplantation genetic technologies: Ethical, social and public policy aspects. Amsterdam: Academic Press, Elsevier (2019). p. 43–52. doi: 10.1016/B978-0-12-816468-6.00006-0

8. Carvalho F, Coonen E, Goossens V, Kokkal G, Rubio C, Meijer-Hoogeveen M, et al. ESHREPGT consortium good practice recommendations for the organisation of PGT. Hum Reprod Open. (2020) 2020(3):hoaa021. doi: 10.1093/hropen/hoaa021

PubMed Abstract | Crossref Full Text | Google Scholar

9. ESHRE PGT-SR/PGT-A Working Group, Coonen E, Rubio C, Christopikou D, Dimitriadou E, Gonta J, et al. ESHRE PGT consortium good practice recommendations for the detection of structural and numerical chromosomal aberrations. Hum Reprod Open. (2020) 2020(3):hoaa017. doi: 10.1093/hropen/hoaa017

PubMed Abstract | Crossref Full Text | Google Scholar

10. ESHRE PGT-M Working Group, Carvalho F, Moutou C, Dimitriadou E, Dreesen J, Giménez C, et al. ESHRE PGT consortium good practice recommendations for the detection of monogenic disorders. Hum Reprod Open. (2020) 2020 (3):hoaa018. doi: 10.1093/hropen/hoaa018

PubMed Abstract | Crossref Full Text | Google Scholar

11. Forman EJ, Tao X, Ferry KM, Taylor D, Treff NR, Scott RT Jr. Single embryo transfer with comprehensive chromosome screening results in improved ongoing pregnancy rates and decreased miscarriage rates. Hum Reprod. (2012) 27(4):1217e22. doi: 10.1093/humrep/des020

Crossref Full Text | Google Scholar

12. Handyside AH, Kontogianni EH, Hardy K, Winston RM. Pregnancies from biopsied human preimplantation embryos sexed by Y-specific DNA amplification. Nature. (1990) 344:768–70. doi: 10.1038/344768a0

PubMed Abstract | Crossref Full Text | Google Scholar

13. Fiorentino F, Kahraman S, Karadayi H, Biricik A, Sertyel S, Karlikaya G, et al. Short tandem repeats haplotyping of the HLA region in preimplantation HLA matching. Eur J Hum Genet. (2005) 13:953–8. doi: 10.1038/sj.ejhg.5201435

PubMed Abstract | Crossref Full Text | Google Scholar

14. Alfarawati S, Fragouli E, Colls P, Wells D. First births after preimplantation genetic diagnosis of structural chromosome abnormalities using comparative genomic hybridization and microarray analysis. Hum Reprod. (2011) 26:1560–74. doi: 10.1093/humrep/der068

PubMed Abstract | Crossref Full Text | Google Scholar

15. Treff NR, Fedick A, Tao X, Devkota B, Taylor D, Scott RT Jr. Evaluation of targeted next-generation sequencing-based preimplantation genetic diagnosis of monogenic disease. Fertil Steril. (2013) 99:1377–84. doi: 10.1016/j.fertnstert.2012.12.018

PubMed Abstract | Crossref Full Text | Google Scholar

16. Handyside AH, Harton GL, Mariani B, Thornhil AR, Afara N, Shaw MA, et al. Karyomapping: a universal method for genome wide analysis of genetic disease based on mapping crossovers between parental haplotypes. J Med Genet. (2010) 47:651–8. doi: 10.1136/jmg.2009.069971

PubMed Abstract | Crossref Full Text | Google Scholar

17. Yan L, Huang L, Xu L, Huang J, Ma F, Zhu X, et al. Live births after simultaneous avoidance of monogenic diseases and chromosome abnormality by next-generation sequencing with linkage analyses. Proc Natl Acad Sci U S A. (2015) 112:15964–9. doi: 10.1073/pnas.1523297113

PubMed Abstract | Crossref Full Text | Google Scholar

18. Rycke MD, Berckmoes V. Preimplantation genetic testing for monogenic disorders. Genes (Basel). (2020) 11:871–86. doi: 10.3390/genes11080871

PubMed Abstract | Crossref Full Text | Google Scholar

19. Schlieben LD, Prokisch H. The dimensions of primary mitochondrial disorders. Front Cell Dev Biol. (2020) 8:600079. doi: 10.3389/fcell.2020.600079

PubMed Abstract | Crossref Full Text | Google Scholar

20. Smeets HJ, Sallevelt SC, Dreesen JC, de Die-Smulders CE, de Coo IF. Preventing the transmission of mitochondrial DNA disorders using prenatal or preimplantation genetic diagnosis. Ann N Y Acad Sci. (2015) 1350:29–36. doi: 10.1111/nyas.12866

PubMed Abstract | Crossref Full Text | Google Scholar

21. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. 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(5):405–24. doi: 10.1038/gim.2015.30

PubMed Abstract | Crossref Full Text | Google Scholar

22. Bean LJH, Funke B, Carlston CM, Gannon JL, Kantarci S, Krock BL, et al. Diagnostic gene sequencing panels from design to report-a technical standard of the American college of medical genetics and genomics (ACMG). Genet Med. (2020) 22:453–61. doi: 10.1038/s41436-019-0666-z

PubMed Abstract | Crossref Full Text | Google Scholar

23. Masson E, Zou WB, Génin E, Cooper DN, Le Gac G, Fichou Y, et al. Expanding ACMG variant classification guidelines into a general framework. Hum Genomics. (2022) 16(1):31. doi: 10.1186/s40246-022-00407-x

PubMed Abstract | Crossref Full Text | Google Scholar

24. Navidi W, Arnheim N. Using PCR in preimplantation genetic disease diagnosis. Hum Reprod. (1991) 6:836–49. doi: 10.1093/oxfordjournals.humrep.a137438

PubMed Abstract | Crossref Full Text | Google Scholar

25. Liu J, Lissens W, Silber SJ, Devroey P, Liebaers I, Van Steirteghem A. Birth after preimplantation diagnosis of the cystic fibrosis delta F508 mutation by polymerase chain reaction in human embryos resulting from intracytoplasmic sperm injection with epididymal sperm. JAMA. (1994) 272:1858–60. doi: 10.1001/jama.1994.03520230068040

PubMed Abstract | Crossref Full Text | Google Scholar

26. Ao A, Handyside A, Winston RM. Preimplantation genetic diagnosis of cystic fibrosis (delta F508). Eur J Obstet Gynecol Reprod Biol. (1996) 65:7–10. doi: 10.1016/0028-2243(95)02294-3

PubMed Abstract | Crossref Full Text | Google Scholar

27. Morsy M, Takeuchi K, Kaufmann R, Veeck L, Hodgen GD, Beebe SJ. Preclinical models for human pre-embryo biopsy and genetic diagnosis. II. Polymerase chain reaction amplification of deoxyribonucleic acid from single lymphoblasts and blastomeres with mutation detection. Fertil Steril. (1992) 57:431–8. doi: 10.1016/S0015-0282(16)54859-6

PubMed Abstract | Crossref Full Text | Google Scholar

28. Farrell PM, Rock MJ, Baker MW. The impact of the CFTR gene discovery on cystic fibrosis diagnosis, counseling, and preventive therapy. Genes (Basel). (2020) 811(4):401. doi: 10.3390/genes11040401

Crossref Full Text | Google Scholar

29. Chauhan W, Fatma R, Zaka-Ur-Rab Z, Afzal M. Direct sequencing of β-globin gene reveals a rare combination of two exonic and two intronic variants in a β-thalassemia major patient: a case report. J Med Case Rep. (2022) 916(1):362. doi: 10.1186/s13256-022-03605-2

Crossref Full Text | Google Scholar

30. Galanello R, Origa R. Beta-thalassemia. Orphanet J Rare Dis. (2010) 5:11. doi: 10.1186/1750-1172-5-11

PubMed Abstract | Crossref Full Text | Google Scholar

31. Crugnola E, Gobbetti A, Fiandanese N, Filippini G. P–562 PGT-M with de novo mutations: how to deal with it? Hum Reprod. (2021) 36(1):i393. doi: 10.1093/humrep/deab130.561

Crossref Full Text | Google Scholar

32. Valdés-Martínez OH, García-Luna SM, Oceguera-Palao LC, Villarreal-Pineda L, Sordia-Hernández LH, de la Fuente Cortez B, et al. Embryo selection for a carrier of an early-onset Alzheimer’s disease-associated mutation in the PSEN1 gene. J Prev Alzheimers Dis. (2023) 10(1):144–7. doi: 10.14283/jpad.2023.4

Crossref Full Text | Google Scholar

33. Ferreira D, Carvalho B, Neto AP, Silva J, Póvoa AM, Barros A, et al. Preimplantation genetic testing for Huntington disease: the perspective of one Portuguese center. Porto Biomed J. (2019) 44(5):e48. doi: 10.1097/j.pbj.0000000000000048

Crossref Full Text | Google Scholar

34. Piyamongkol S, Makonkawkeyoon K, Shotelersuk V, Sreshthaputra O, Pantasri T, Sittiwangkul R, et al. Pre-implantation genetic testing for Marfan syndrome using mini-sequencing. J Obstet Gynaecol. (2022) 42(7):2846–52. doi: 10.1080/01443615.2022.2109957

PubMed Abstract | Crossref Full Text | Google Scholar

35. Thornhill AR, Snow K. Molecular diagnostics in preimplantation genetic diagnosis. J Mol Diagn. (2002) 4:11–29. doi: 10.1016/S1525-1578(10)60676-9

PubMed Abstract | Crossref Full Text | Google Scholar

36. Thornhill AR, de Die Smulders CE, Geraedts JP, Harper JC, Harton GL, Lavery SA, et al. ESHRE PGD consortium ‘best practice guidelines for clinical preimplantation genetic diagnosis (PGD) and preimplantation genetic screening (PGS). Hum Reprod. (2005) 20:35–48. doi: 10.1093/humrep/deh579

PubMed Abstract | Crossref Full Text | Google Scholar

37. Patel N, Bhadarka HK, Vaniawala S, Patel A. A successful case for deselection of albino embryo and live birth of albinism-free healthy baby followed by PGT-M. J Hum Reprod Sci. (2020) 13(3):245–8. doi: 10.4103/jhrs.JHRS_38_19

PubMed Abstract | Crossref Full Text | Google Scholar

38. Sallevelt SCEH, de Koning B, Szklarczyk R, Paulussen ADC, de Die-Smulders CEM, Smeets HJM. A comprehensive strategy for exome-based preconception carrier screening. Genet Med. (2017) 19(5):583–92. doi: 10.1038/gim.2016.153

PubMed Abstract | Crossref Full Text | Google Scholar

39. Ren Y, Lian Y, Yan Z, Zhai F, Yang M, Zhu X, et al. Clinical application of an NGS-based method in the preimplantation genetic testing for Duchenne muscular dystrophy. J Assist Reprod Genet. (2021) 38:1979–86. doi: 10.1007/s10815-021-02126-z

PubMed Abstract | Crossref Full Text | Google Scholar

40. Fu Y, Shen X, Wu H, Chen D, Zhou C. Preimplantation genetic testing for monogenic disease of spinal muscular atrophy by multiple displacement amplification: 11 unaffected livebirths. Int J Med Sci. (2019) 16(9):1313–9. doi: 10.7150/ijms.32319

PubMed Abstract | Crossref Full Text | Google Scholar

41. Chen M, Chang SP, Ma GC, Lin WH, Chen HF, Chen SU, et al. Preimplantation genetic diagnosis of hemophilia A. Thromb J. (2016) 14(1):33. doi: 10.1186/s12959-016-0098-9

PubMed Abstract | Crossref Full Text | Google Scholar

42. Iglesias A, Anyane-Yeboa K, Wynn J, Wilson A, Truitt Cho M, Guzman E, et al. The usefulness of whole-exome sequencing in routine clinical practice. Genet Med. (2014) 16(12):922–31. doi: 10.1038/gim.2014.58

PubMed Abstract | Crossref Full Text | Google Scholar

43. Doroftei B, Ilie OD, Anton N, Armeanu T, Ilea C. A mini-review regarding the clinical outcomes of in vitro fertilization (IVF) following Pre-implantation genetic testing (PGT)-next generation sequencing (NGS) approach. Diagnostics. (2022) 12(8):1911. doi: 10.3390/diagnostics12081911

PubMed Abstract | Crossref Full Text | Google Scholar

44. Trieutien S, Vu Van T, Tran Ngoc Thao M, Trinh The S, Tran Van K, Nguyen Thanh T, et al. Preimplantation genetic diagnosis for DEB by detecting a novel family-specific COL7A1 mutation in Vietnam. Appl Clin Genet. (2021) 914:467–72. doi: 10.2147/TACG.S344107

Crossref Full Text | Google Scholar

45. Yeates L, McDonald K, Burns C, Semsarian C, Carter S, Ingles J. Decision-making and experiences of preimplantation genetic diagnosis in inherited heart diseases: a qualitative study. Eur J Hum Genet. (2022) 30(2):187–93. doi: 10.1038/s41431-021-00963-1

PubMed Abstract | Crossref Full Text | Google Scholar

46. Neumann A, Alcantara-Ortigoza MA, González-Del Angel A, Zarate Díaz NA, Santana JS, Porchia LM, et al. Whole-genome amplification/preimplantation genetic testing for propionic acidemia of successful pregnancy in an obligate carrier Mexican couple: a case report. World J Clin Cases. (2021) 9(29):8797–803. doi: 10.12998/wjcc.v9.i29.8797

PubMed Abstract | Crossref Full Text | Google Scholar

47. Porto A, Caffrey RG, Crowley-Matoka M, Spencer S, Li M, Propst L. Offering pre-implantation genetic testing for monogenic disorders (PGT-M) for conditions with reduced penetrance or variants of uncertain significance: ethical insight from U.S. laboratory genetic counselors. J Genet Couns. (2021) 1(1):261–8. doi: 10.1002/jgc4.1482

Crossref Full Text | Google Scholar

48. Madon P. Genetic counseling and Pre-PGT workup. In: Ahuja M, Malhotra N, Loomba P, editors. ISARCON 2017 manual of preimplantation genetic diagnosis and screening, vitrification of gametes, embryos and tissues. New Delhi: Jaypee (2017). p. 32–9.

49. Unsal E, Aktaş Y, Uner O, BaltacI A, Ozcan S, Turhan F, et al. Successful application of preimplantation genetic diagnosis for leigh syndrome. Fertil Steril. (2008) 90(5):2017.e11–3. doi: 10.1016/j.fertnstert.2008.07.023

PubMed Abstract | Crossref Full Text | Google Scholar

50. Poulton J, Finsterer J, Yu-Wai-Man P. Genetic counseling for maternally inherited mitochondrial disorders. Mol Diagn Ther. (2017) 21(4):419–29. doi: 10.1007/s40291-017-0279-7

PubMed Abstract | Crossref Full Text | Google Scholar

51. Smeets HJM, Sallevelt SCEH, Herbert M. Reproductive options in mitochondrial disease. Handb Clin Neurol. (2023) 194:207–28. doi: 10.1016/B978-0-12-821751-1.00004-X

PubMed Abstract | Crossref Full Text | Google Scholar

52. Parikh FR, Naik DJ, Naik NJ, Sanap RR, Madon PF, Sanap MV, et al. First successful twin delivery in India after preimplantation genetic diagnosis for BRCA1 mutation. Indian J Appl Res. (2018) 8(8):3–4. doi: 10.36106/ijar

Crossref Full Text | Google Scholar

53. Yahalom C, Macarov M, Lazer-Derbeko G, Altarescu G, Imbar T, Hyman JH, et al. Preimplantation genetic diagnosis as a strategy to prevent having a child born with an heritable eye disease. Ophth Genet. (2018) 39(4):450–6. doi: 10.1080/13816810.2018.1474368

Crossref Full Text | Google Scholar

54. Vriesen N, Carmany EP, Natoli JL. Clinical outcomes of preimplantation genetic testing for hereditary cancer syndromes: a systematic review. Prenat Diagnos. (2022) 42(2):201–11. doi: 10.1002/pd.6084

Crossref Full Text | Google Scholar

55. Kuliev K, Pakhalchuk T, Prokhorovich M, Rechitsky S. Preimplantation genetic testing for inherited cancer predisposition. Adv Can Res Clin Imaging. (2021) 3(2):ACRCI.MS.ID.000557. doi: 10.33552/ACRCI.2021.03.000557

Crossref Full Text | Google Scholar

56. Daar J, Benward J, Collins L, Davis J, Davis O, Francis L, et al. Use of preimplantation genetic testing for monogenic defects (PGT-M) for adult-onset conditions: an ethics committee opinion. Fertil Steril. (2018) 109(6):989–92. doi: 10.1016/j.fertnstert.2018.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

57. Ayrignac X, Carra-Dallière C, Marelli C, Taïeb G, Labauge P. Adult-onset genetic central nervous system disorders masquerading as acquired neuro inflammatory disorders: a review. JAMA Neurol. (2022) 79(10):1069m78. doi: 10.1001/jamaneurol.2022.2141

Crossref Full Text | Google Scholar

58. Gilchrist DM. Medical genetics: 3. An approach to the adult with a genetic disorder. CMAJ. (2002) 29167(9):1021–9. PMID: 12403743; PMCID: PMC134180

PubMed Abstract | Google Scholar

59. Lewis CM, Vassos E. Polygenic risk scores: from research tools to clinical instruments. Genome Med. (2020) 12:44. doi: 10.1186/s13073-020-00742-5

PubMed Abstract | Crossref Full Text | Google Scholar

60. Treff NR, Eccles J, Marin D, Messick E, Lello L, Gerber J, et al. Preimplantation genetic testing for polygenic disease relative risk reduction: evaluation of genomic index performance in 11,883 adult sibling pairs. Genes (Basel). (2020) 11:648. doi: 10.3390/genes11060648

PubMed Abstract | Crossref Full Text | Google Scholar

61. Treff NR, Marin D, Lello L, Hsu S, Tellier LC. Preimplantation genetic testing: preimplantation genetic testing for polygenic disease risk. Reproduction. (2020) 160:A13–7. doi: 10.1530/REP-20-0071

PubMed Abstract | Crossref Full Text | Google Scholar

62. Rubio C, Simón C. Embryo genetics. Genes (Basel). (2021) 12:118. doi: 10.3390/genes12010118

PubMed Abstract | Crossref Full Text | Google Scholar

63. Griffin DK, Gordon AT. Preimplantation testing for polygenic disease (PGT-P): brave new world or mad pursuit? DNA. (2023) 3(2):104–8. doi: 10.3390/dna3020008

Crossref Full Text | Google Scholar

64. Siermann M, Tšuiko O, Vermeesch JR, Raivio T, Borry P. A review of normative documents on preimplantation genetic testing: recommendations for PGT-P. Genet Med. (2022) 24(6):1165–75, ISSN 1098-3600. doi: 10.1016/j.gim.2022.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

65. Tsui DW, Chiu RW, Lo YD. Epigenetic approaches for the detection of fetal DNA in maternal plasma. Chimerism. (2010) 1(1):30–5. doi: 10.4161/chim.1.1.12439

PubMed Abstract | Crossref Full Text | Google Scholar

66. Poli M, Girardi L, Fabiani M, Moretto M, Romanelli V, Patassini C, et al. Past, present, and future strategies for enhanced assessment of embryo’s genome and reproductive competence in women of advanced reproductive age. Front Endocrinol (Lausanne). (2019) 1910:154. doi: 10.3389/fendo.2019.00154

Crossref Full Text | Google Scholar

67. Kakourou G, Vrettou C, Moutafi M, Traeger-Synodinos J. Pre-implantation HLA matching: the production of a savior child. Best Pract Res Clin Obstet Gynecol. (2017) 44:76–89. doi: 10.1016/j.bpobgyn.2017.05.008

Crossref Full Text | Google Scholar

68. Verlinsky Y, Rechitsky S, Schoolcraft W, Strom C, Kuliev A. Preimplantation diagnosis for fanconi anemia combined with HLA matching. JAMA. (2001) 285(24):3130–3. doi: 10.1001/jama.285.24.3130

PubMed Abstract | Crossref Full Text | Google Scholar

69. Athalye AS, Naik DJ, Sanap RR, Madon PF, Warang DJ, Parikh FR. Birth of a savior sibling after preimplantation genetic testing for beta-thalassemia, HLA haplotyping and aneuploidy screening. Polymorph. (2021) 7:64–72.

Google Scholar

70. De Rycke M, De Vos A, Belva F, Berckmoes V, Bonduelle M, Buysse A, et al. Preimplantation genetic testing with HLA matching: from counseling to birth and beyond. J Hum Genet. (2020) 65:445–54. doi: 10.1038/s10038-020-0732-z

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: PGT, genetic counseling, preimplantation genetic testing, PGT-M, monogenic disorders, GC for PGT-M, Mendelian disorders

Citation: Parikh F, Athalye A, Madon P, Khandeparkar M, Naik D, Sanap R and Udumudi A (2023) Genetic counseling for pre-implantation genetic testing of monogenic disorders (PGT-M). Front. Reprod. Health 5:1213546. doi: 10.3389/frph.2023.1213546

Received: 28 April 2023; Accepted: 4 December 2023;
Published: 15 December 2023.

Edited by:

Bianca Bianco, Faculdade de Medicina do ABC, Brazil

Reviewed by:

Bruno Coprerski, Igenomix, Brazil
Alexandre Dias, Paulista University, Brazil
Monica Lipay, Faculty of Medicine of Jundiaí, Brazil

© 2023 Parikh, Athalye, Madon, Khandeparkar, Naik, Sanap and Udumudi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Firuza Parikh frparikh@gmail.com

These authors have contributed equally to this work and share first authorship

These authors have contributed equally to this work and share senior authorship

§These authors have contributed equally to this work and share last authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.