Signed in as:
filler@godaddy.com
Signed in as:
filler@godaddy.com
Hereditary cancer genetic testing allows the identification of genetic variants that can increase the risk of developing cancer. Early identification of cancer predisposition genes can lead to better clinical management of the disease in the future.
Testing technologies range from chromosomal analysis to whole exome or whole genome analysis. To diagnose cancer type cancer or to evaluate the risk of particular cancer type, single gene or multiple genes (gene panel) analysis are commonly used.
Dying cancer cells release their mutated DNA into the bloodstream, enabling determination of mutation type and relative tumor volume. In this non-invasive technique, by capturing and sequencing the tumor-derived cell-free DNA (cfDNA), tumor genomic profile can be reconstructed without needing to perform a biopsy of the tumor.
This process is also called liquid biopsy, which is designed to sequence regions of oncogenes. Liquid biopsy Facilitates early detection of emergent genetic alterations that can be associated with resistance to therapy during cancer progression.
Cancer is initiated from genetic alterations (changes in the DNA or mutations) that can either be inherited from parents or acquired sporadically during a person’s lifetime.
Over time, the mutations accumulate resulting in the production of abnormal proteins which can cause healthy cells to become cancerous and can be responsible for cancer progression as well as therapy resistance.
A set of a wide range of tests offered to understand the molecular and genetic basis of leukemias in order to make more informed treatment-related decisions.
A combination of various techniques and multiple markers tested by each technique that provides a comprehensive understanding of a patient’s leukemia to the treating clinician.
Cancer is a genetic disease characterized by the rapid and uncontrolled growth of abnormal cells. It is one of the leading causes of death worldwide. Cancer is a multi- stage process which starts from genetic alterations in the DNA of normal cells, known as mutations. Accumulation of genetic alterations causes the now cancerous cells to multiply and form a mass, known as a tumor, which can grow and spread to other parts of the body.
90-95% of cancer types occur sporadically without an inherited genetic cause and 5-10% of cancer cases can run in families (inherited).
Inherited mutations (genetic variants) can exist from birth and can be passed down from parents to their children, increasing the risk of developing cancer in the future.
Identifying inherited variants and estimating the risk for cancer development provides the opportunity to identify the most appropriate preventive measures, offer better clinical management and/or undergo timely routine monitoring.
Everyone inherits one copy of the gene pair from the mother and another one from the father. When a genetic mutation is present in the germ cells (which means eggs or sperm from parents) and inherited to the individuals from birth, it is referred to as germline mutation.
Not all germline mutation carriers will develop cancer. However, carrying a germline mutation in a oncogene, tumor suppressor or DNA repair genes may increase the likelihood of developing cancer in a lifetime.
A somatic mutation, or acquired mutation, happens in somatic (body) cells instead of germ cells and will not pass to offspring. A somatic mutation is frequently caused by environmental factors, such as smoking or exposure to carcinogens.
Classes of Cancer Genes: genes related to Cancer Development are of different functional classes:
In most cases, cancer is not caused by a single gene mutation, it usually takes several mutations impacting multiple genes to develop cancer.
Understanding the genetics of cancer may help understand the diagnosis, treatment, and prevention of the disease.
Families or individuals with the following cancers or syndromes shall be tested for risk of hereditary cancer:
Please reach us at contact@genatechs.com if you cannot find the test you are searching for.
AIP, ALK, APC, AR, ATM, BAP1, BARD1, BLM, BMPR1A, BRCA1, BRCA2, BRIP1, BUB1B, CD82, CDC73, CDH1, CDK4, CDKN1C, CDKN2A, CEBPA, CEP57, CHEK2, CYLD, DDB2, DICER1, DIS3L2, EGFR, ELAC2, ENG, EPCAM, ERCC2, ERCC3, ERCC4, ERCC5, EXT1, EXT2, EZH2, FANCA, FANCB, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCL, FANCM, FH, FLCN, GATA2, GPC3, HRAS, KIT, MAX, MEN1, MET, MLH1, MLH3, MRE11A, MSH2, MSH3, MSH6, MSR1, MUTYH, MXI1, NBN, NF1, NF2, NSD1, PALB2, PHOX2B, PMS1, PMS2, PRF1, PRKAR1A, PTCH1, PTEN, RAD50, RAD51C, RAD51D, RB1, RECQL4, RET, RHBDF2, RNASEL, RUNX1, SBDS, SDHAF2, SDHB, SDHC, SDHD, SLX4, SMAD4, SMARCB1, STK11, SUFU, TGFBR2, TMEM127, TP53, TSC1, TSC2, VHL, WRN, WT1, XPA, XPC
Single Gene
Reflex MLPA for BRCA1 & BRCA 2 / Mismatch repair genes
(SNVs, small INDELs and CNVs) including BRCA1 and BRCA2)
ABRAXAS1, AIP, ALK, APC, AR, ATM, AXIN2, BAP1, BARD1, BLM, BMPR1A, BRCA1, BRCA2, BRIP1, BUB1B, CBL, CDC73, CDH1, CDK4, CDKN1B, CDKN1C, CDKN2A, CHEK2, CTNNA1, CYLD, DDB2, DICER1, DIS3L2, ELAC2, EPCAM, ERCC2, ERCC3, ERCC4, ERCC5, EXT1, EXT2, FAN1, FANCA, FANCB, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCL, FANCM, FH, FLCN, GALNT12, GATA2, GPC3, HOXB13, HRAS, KIF1B, KIT, LZTR1, MAX, MEN1, MET, MITF, MLH1, MLH3, MRE11, MSH2, MSH3, MSH6, MSR1, MUTYH, NBN, NF1, NF2, NTHL1, PALB2, PAX5, PDGFRA, PHOX2B, PMS1, PMS2, POLD1, POLE, POT1, PRKAR1A, PRSS1, PTCH1, PTCH2, PTEN, RAD50, RAD51C, RAD51D, RB1, RECQL, RECQL4, RET, RHBDF2, RINT1, RNASEL, RNF43, RUNX1, SBDS, SDHA, SDHAF2, SDHB, SDHC, SDHD, SLC45A2, SLX4, SMAD4, SMARCB1, SMARCE1, SRGAP1, STK11, SUFU, TERT, TGFBR2, TMEM127, TP53, TSC1, TSC2, TYR, VHL, WRN, WT1, XPA, XPC, XRCC2, XRCC3, GREM2, SCG5
158 genes including BRCA1 and BRCA2 Plus deletion/duplication analysis of 30 genes including BRCA1, BRCA2 and HRR genes
APC, ATM, BAP1, BARD1, BMPR1A, BRCA1, BRCA2, BRIP1, CDH1, CDK4, CDKN2A, CHEK2, EPCAM, GREM2, MITF, MLH1, MSH2, MSH6, MUTYH, NBN, PALB2, PMS2, POLE, PTEN, RAD51C, RAD51D, SCG5, SMAD4, STK11, TP53
AIP, ALK, APC, AR, ATM, BAP1, BARD1, BLM, BMPR1A, BRCA1, BRCA2, BRIP1, BUB1B, CD82, CDC73, CDH1, CDK4, CDKN1C, CDKN2A, CEBPA, CEP57, CHEK2, CYLD, DDB2, DICER1, DIS3L2, EGFR, ELAC2, ENG, EPCAM, ERCC2, ERCC3, ERCC4, ERCC5, EXT1, EXT2, EZH2, FANCA, FANCB, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCL, FANCM, FH, FLCN, GATA2, GPC3, HRAS, KIT, MAX, MEN1, MET, MLH1, MLH3, MRE11A, MSH2, MSH3, MSH6, MSR1, MUTYH, MXI1, NBN, NF1, NF2, NSD1, PALB2, PHOX2B, PMS1, PMS2, PRF1, PRKAR1A, PTCH1, PTEN, RAD50, RAD51C, RAD51D, RAD54L, RB1, RECQL4, RET, RHBDF2, RNASEL, RUNX1, SBDS, SDHAF2, SDHB, SDHC, SDHD, SLX4, SMAD4, SMARCB1, STK11, SUFU, TGFBR2, TMEM127, TP53, TSC1, TSC2, VHL, WRN, WT1, XPA, XPC; BRCA1 & BRCA2 (MLPA)
MLH1, MSH2, MSH6, PMS1, EPCAM, MSH3, MLH3, PMS2
Single Gene
Single Gene
Single Gene
Single Gene
Cancer patients who fall in one of the following categories can get tested with liquid
biopsy:
Genatechs provides NGS based Liquid biopsy testing panels. The advantages of this test using NGS technology are as follows:
Please reach us at contact@genatechs.com if you cannot find the test you are searching for.
Screens for the presence of oncogenic driver mutations in four key genes: EGFR, KRAS, NRAS and BRAF.
HOTSPOT mutations in EGFR (Exons 18, 19, 20, 21; includes T790M), KRAS (Exons 2, 3, 4), NRAS (Exons 2, 3, 4), BRAF (V600E)
110+ genes for all solid tumors detects SNVs & InDels
110+ genes for all solid tumors detects SNVs, InDels & Fusions
Comprehensive Genomic Profiling of Solid Tumors from Blood | High precision multibiomarker test | SNVs & InDels - 523 genes I CNVs - 59 genes I Fusion - 23 genes | MSI | TMB
24 genes for NSCLC (SNVs, InDels & Fusions)
(15 Homologous Recombination Repair pathway, including BRCA1 & BRCA2 genes)
Includes HOTSPOT mutations in EGFR (Exons 18, 19, 20, 21; includes T790M)
HOTSPOT mutations in EGFR (T790M - indicating resistance to first & second
gen TKI and C797S indicating resistance to third gen TKIs) Primary use in Adenocarcinoma – Lung
Inclusive genes: ABL1, AKT1, ALK, APC, ATM, BRAF (inclusive of v600E), CDH1, CDKN2A, CSF1R, CTNNB1, DDR2, DNMT3A, EGFR, ERBB2, ERBB4, EZH2, FBXW7, FGFR1, FGFR2, FGFR3, FLT3, FOXL2, GNA11, GNAQ, GNAS, HNF1A, HRAS, IDH1, IDH2, JAK2, JAK3, KDR, KIT, KRAS, MAP2K1/MEK1, MET, MLH1, MPL, MSH6, NOTCH1, NPM1, NRAS, PDGFRA, PIK3CA, PTEN, PTPN11, RB1, RET, SMAD4, SMARCB1, SMO, SRC, STK11, TP53, TSC1, VHL
Except for TP53 only specific hotspots are covered in the aforementioned genes, that is, those which are therapeutically relevant.
Specific exonic coverage will be provided on individual reports
Contact for more information
Tumor profiling is a laboratory-based method which identifies mutations, genomic biomarkers (such as Tumor Mutational Burden, TMB; and Microsatellite Instability, MSI) as well as other molecules including proteins which can be responsible for cancer development and resistance to treatment.
Different molecular profiling techniques, such as immunohistochemistry and genetic testing, can help in deep molecular characterization of the tumors either at the protein or the gene level.
Tumor profiling allows the investigation of genomic mechanisms involved in tumor formation, including microsatellite instability and fusion genes, as well as enabling investigation of tumor mutation burden analysis.
Microsatellites are repeated sections of DNA, 1-6bp long, that are found throughout the entire genome and account for approximately 3% of it. Due to their repeated sequences, microsatellites are prone to a high mutation rate. Microsatellite instability (MSI) is a cause of unique molecular alterations and hypermutated phenotypes. It is triggered by an impaired DNA mismatch repair (MMR) system, which frequently results from germline or somatic mutations or promoter hypermethylation of genes in the DNA MMR system, such as MLH1, MLH2, MSH6, and PMS2.
Determined as a predictive biomarker and can support the selection of patients who may benefit from immune checkpoint inhibitor (ICI) therapy. TMB is defined as the number of somatic coding, non-synonymous variants in the tumor genome per megabase and is associated with the development of neoantigens that trigger antitumor immunity.
Occur across a wide spectrum of tumor types. Gene fusions arise as a result of genomic rearrangements, including chromosomal inversions, interstitial deletions, duplications, or translocations, and can drive both the development and progression of cancer. Sequencing efforts have identified rare oncogenic fusions across several forms of cancer. Many of these fusions have proven to be viable targets or are the subject of promising ongoing research.
Histopathological examination and genetic testing can determine the tumor profile and guide management and treatment plans.
Solid tumors represent approximately 90% of adult cancers. In up to 40% of patients, complex genomic alterations are identified, which can serve as biomarkers to predict response to specific therapies and/or prognosis.
Your physician can choose from individual targeted therapy tests, gene panels or a single comprehensive gene panel.
We offer
Please reach us at contact@genatechs.com if you cannot find the test you are searching for.
It is a next generation sequencing (NGS) assay consisting of DNA and RNA based testing which detects Tumor mutations {single nucleotide variants (SNVs), insertion-deletion mutations (InDels), and copy number variants (CNVs)}, fusions, tumor mutation burden (TMB), microsatellite instability (MSI) and PD-L1 expression in multiple solid tumors.
77 genes SNVs, Indels, CNVs and Fusions
231 genes SNVs, Indels, CNVs
Single Gene
Single Gene
~400 genes SNVs, Indels, CNVs, and Fusions
231 genes SNVs, Indels, CNVs and TMB
~400 genes SNVs, Indels, CNVs, Fusions and TMB
~400 genes SNVs, Indels, CNVs, Fusions, TMB and MSI
ATK1, FGFR1, AR, FGFR2, BRCA1, PIK3CA, BRCA2, PTEN, ERBB2, PD-L1, ESR1, and PGR
BRCA1, BRCA2, KRAS, PDGFRA, FOXL2, TP53, ATM, BARD1, BRIP1, CDK12, CHEK1, CHEK2, FANCL, PALB2, PPP2R2A, RAD51B, RAD51C, RAD51D, and RAD54L
POLE, MLH1, MSH2, PMS2, MSH6, TP53, ERBB2, and ESR1
AKT1, MSH2, BRAF, MSH6, HRAS, NRAS, KRAS, PIK3CA, MET, PMS2, MLH1, PTEN, SMAD4, and Her2 Amplification
ALK, BRAF, BRCA1, BRCA2, ERBB2, KRAS, NRG1, PALB2, ROS1
ATM, BRCA1, BRCA2, MLH1, MSH2, PMS2, MSH6, AR, BARD1, BRIP1, CDK12, CHEK1, CHEK2, FANCL, PALB2, PPP2R2A, RAD51B, RAD51C, RAD51D, and RAD54L
AKT1, ALK, BRAF, DDR2, EGFR, ERBB2, FGFR1, FGFR2, FGFR3, KRAS, MAP2K1, MET, NRAS, PIK3CA, PTEN, RET, TP53, PD-L1*, and Her2 Amplification
NTRK1, NTRK2, NTRK3, TMB, MSI, and PD-L1*
*Performed by IGC (SP263/SP142/22c3)
Cancer specific Molecular profiling (Combos and panels)
Details
Details
Details
Details
Details
Details
Details
Details
Tests cover markers that are of diagnostic, prognostic, and therapeutic importance. Based on individual leukemia subtype there are various markers to be evaluated according to a work-up by bodies like World Health Organization (WHO) and American Society for Hematology (ASH). The diagnostic markers can be tested during the presentation of the disease, whereas prognostic and therapeutic markers can be tested while making therapeutic decisions.
A set of a wide range of tests offered to understand the molecular and genetic basis of leukemias in order to make more informed treatment-related decisions.
A combination of various techniques and multiple markers tested by each technique that provides a comprehensive understanding of a patient’s leukemia to the treating clinician.
We use cookies to analyze website traffic and optimize your website experience. By accepting our use of cookies, your data will be aggregated with all other user data.