fbpx
Wikipedia

T-cell acute lymphoblastic leukemia

T-cell acute lymphoblastic leukemia (T-ALL) is a type of acute lymphoblastic leukemia with aggressive malignant neoplasm of the bone marrow.[6] Acute lymphoblastic leukemia (ALL) is a condition where immature white blood cells accumulate in the bone marrow, subsequently crowding out normal white blood cells[7] and create build-up in the liver, spleen, and lymph nodes.[8] The two most common types of ALL are B-lymphocytes and T-lymphocytes, where the first protects the body against viruses and bacteria through antibody production which can directly destroy target cells or trigger others to do so, whilst the latter directly destroy bacteria or cells infected with viruses.[9] Approximately 20% of all ALL patients are categorized specifically to suffer from T-ALL and it is seen to be more prevalent in the adult population in comparison to children, with incidences[spelling?] shown to diminish with age.[6][10] Amongst T-ALL cases in the pediatric population, a median onset of age 9 has been identified and the disease is particularly prominent amongst adolescents.[6] The disease stems from cytogenic and molecular abnormalities, resulting in disruption of developmental pathways controlling thymocyte development, tumor suppressor development, and alterations in control of cell growth and proliferation.[1] Distinct from adult T-cell leukemia where T-cell lymphotropic virus Type I causes malignant maturation of T-cells, T-ALL is a precursor for lymphoid neoplasm.[6] Its clinical presentation most commonly includes infiltration of the central nervous system (CNS), and further identifies mediastinal mass presence originating from the thymus, along with extramedullary involvement of multiple organs including the lymph node as a result of hyperleukocytosis.

T-cell acute lymphoblastic leukemia
T-lymphoblastic cells of acute leukemia in the bone marrow. In some cases, the cytoplasm is concentrated at one pole of the cell, forming "hand mirror cells".
SpecialtyHaematology, oncology
SymptomsRecurrent infections, unusual or common bleeding and bruising, extreme tiredness, unexplained fever, unexplained weight gain, swollen lymph nodes
Usual onsetMost prevalent in the adult population with incidences[spelling?] diminishing with age. Amongst pediatric population, median onset of age 9. Marked male predominance [1]
CausesCurrently unknown
Diagnostic methodBlood test, bone marrow aspiration,[2] biopsy, CT, MRI, lumbar puncture,[2] genetic testing
TreatmentLong-term chemotherapy,[3] CNS radiation therapy,[1] stem cell transplantation [4]
Prognosis5-Year Event Free Survival: 70%, Overall Survival: 80% [1]
Frequency7% at ages 1-10, 14% at ages 10-15, and 29% at ages 15-18 [5]

Signs and symptoms edit

T-ALL patients may not always experience all the signs and symptoms below. Patients with other medical conditions that are not leukemia may also experience similar symptoms.

  • Recurrent infections due to lack of normal white blood cells (neutrophils)[11]
  • Unusual and/or common bleeding and bruising
  • Extreme tiredness and swellings in the neck (lymph nodes) or the middle of the chest, causing possible facial swelling
  • Unexplained fevers, chills, and/or night sweats
  • Unexplained weight loss and/or loss of appetite
  • Swollen lymph nodes
  • Unexplained skin itch

Clinical manifestations edit

Originating from epigenetic and genetic alterations in immature thymocytes, T-ALL is a highly aggressive and heterogenous disease. Patients often present extensive bone marrow involvement, mediastinal mass, adenopathy, CNS involvement, and splenomegaly.[1] Symptoms can be presented acutely or develop progressively over time. The most common clinical feature amongst patients is the proliferation of malignant clones, hence suppressing normal hematopoiesis, resulting in deficiency of functioning peripheral blood cells (particularly thrombocytes) deficiency.[1]

Risk factors edit

T-ALL is not a contagious nor inherited condition. Its two main risk factors are age and gender.[8] Most cases of leukemia increase with age, with ALL being the main exception, which peaks in children aged 2 to 5 years. T-ALL is seen to be most prevalent in the adult population, but amongst cases in the pediatric population, it is seen to have a median onset of age 9 and is most prominent to adolescents.[6][10] The disease also is marked male predominance with a three-fold increased risk of developing T-ALL in comparison to females. It is currently unclear as to why T-ALL is preferential towards older children and males.[1]

Cytogenetics edit

Basic karyotyping showed structural chromosomal rearrangements in 50-75% of T-ALL patients, primarily inversion and translocations.[1] Diagnostic yield can be substantially increased through further diagnosis through fluorescent in situ hybridization (FISH) and other various molecular technologies, for example single nucleotide polymorphism (SNP) array. The most common structural abnormality is rearrangement of the TCR gene. 95% of T-cell TCR consist of an alpha and beta chain (encoded by TRA and TRB, respectively), where only 5% of T-cell TCR consists of gamma and delta chains (encoded by TRG and TRD, respectively).[4]

Karyotyping showed that TRD and TRB undergo recombination most commonly, whereas TRA is seldom involved and TRG is rarely rearranged. These rearrangements affect the normal process of TCR and could lead to cellular machinery failing to correctly repair recombination-activating RAG protein induced double-strand breaks.[1] All 30 genes known to illegitimately recombine with TCR genes function primarily to regulate epigenetics through roles such as signal transducers, transcription factors (tumor suppressors or oncogenes), cell cycle regulators, or ribosomal proteins.

T-Cell TCR encoded by TRA, TRD, and TRG at chromosome bands 14q11 and 7q34 become malignant in T-ALL patients.[1] The build-up of malignant T-cells in T-ALL are clones with identical T-cell receptor gene arrangements having taken rise from a single cell. The gene rearrangements, as a result of the malignant cell, juxtapose both TCR genes and other critical genes that code for transcription factors. This results in dysregulation of partner gene transcription, which serves as the main cause of leukemogenesis – a multi-step process of induction, development, and progression of leukemic diseases.[1] 20% of all leukemias demonstrate simultaneous rearrangement of these genes.

Pathology edit

Like most cancers, mutations in the DNA begin T-ALL development and lead to loss of function of white blood cells. Different subtypes of leukemia have similarities in their causes, which are a combination of genetics, epigenetic changes, and environmental factors. However, because there are few T-ALL cases in comparison to other subtypes of leukemia, there is currently no clear cause of T-ALL. T-ALL is not contagious nor inherited but specific genetic mutations, commonly including NOTCH1 and CDKN2A, may be passed along which increases susceptibility of T-ALL.[10]

Causes of T-ALL edit

Genetic conditions edit

Some patients may have familial histories with leukemia predispositions which increases risk of developing T-ALL. Li–Fraumeni syndrome is an inherited condition that leads to mutation of TP53, a tumor suppressor gene, which then increases risk of T-ALL. Mutation in gene SPRED1 is also associated with development of T-ALL.

Patients with immature thymocytes in the thymus begins T-ALL development. Furthermore, hereditary conditions such as Down syndrome, neurofibromatosis type 1, ataxia telangiectasia, and Noonan syndrome are associated with higher risk of developing T-ALL.

Radiation exposure edit

Human T-Lymphotropic Virus

Those who have had previous chemotherapy and exposure to radiation may have increased risks of developing T-ALL. CDKN2A is an inherited polymorphism variant that is seen to be associated with development of T-ALL. SR-90 emission from nuclear reactor accidents is also believed to increase risk of developing T-ALL.

Chemical exposure edit

Benzene, a chemical classified as being carcinogenic to humans, is associated with increased risk of T-ALL, as well as other forms of leukemia.[12]

Viruses edit

Human T-lymphotropic virus (HTLV-1) is a retroviral infection that affect white blood cells (T-cells), which may later develop into T-ALL and other subtypes of leukemia.[13]

Diagnosis edit

When doctors are suspicious of a patient potentially suffering from T-ALL after careful examination of background (including medical history, signs, and symptoms), doctors would then conduct tests, procedures, and scans to proceed with diagnosis of T-ALL. Some symptoms and medical history may not be specific enough to diagnose T-ALL, so further testing may be required. Doctors may consider some factors mentioned but would not necessarily conduct all tests possible.[11]

Assessments edit

Blood tests edit

Complete blood count (CBC) is done to test for T-ALL by measuring the different types and maturity of cells in the patient's blood, which allows the donor to determine whether leukemic cells are present in the patient. Additionally, blood tests that show high levels of white blood cells or low levels of red blood cells may also be a sign of T-ALL. Further testing could also help indicate whether T-ALL has affected other organs such as the kidneys as well as the genetic alterations of the disease.

Bone marrow aspiration and biopsy edit

Bone marrow consists of a combination of solid and liquid components. Bone marrow aspiration and biopsies are typically done simultaneously to help determine and confirm the type and severity of T-ALL. Further biopsies such as skin and lymph node biopsies may also need to be done to check for the spread of T-ALL.[2]

X-rays and ultrasound edit

As swollen spleen and lymph nodes are symptoms of T-ALL, X-rays and ultrasound scans, such as CT and MRI, can help confirm the diagnosis. This also provides information on the impact T-ALL has on other organs of the body.

Lumbar puncture edit

To prevent ineffective treatments towards T-cells that have invaded the CNS, lumbar puncture allows doctors to determine whether the treatments will be effective. This also reveals the spread of T-ALL.[2]

 
Lumbar Puncture Positions

Genetic test edit

Genetic testing helps identify chromosomal abnormalities in the patients. This can help identify the genetic mutations and therefore diagnose the specific leukemia subtype.

Staging edit

Normal staging is not used for T-ALL because it is already spread throughout the body when first diagnosed. However, they have their own system of classifying T-ALL cases.[14] First, patterns of gene expression are investigated to define T-ALL. Then, stages of thymic development can be determined by identifying specific expressions in chromosomal abnormalities. This forms the stages of T-ALL cases being either at high or low risk.[8] Patients will then receive the appropriate treatment in respect to whichever class they are in.[14]

Treatment edit

Currently, standard treatment of T-ALL takes the form of long-term chemotherapy and drug intake to prevent or treat side effects associated with low white blood cell count as a result of intensive chemotherapy regimes. The treatment typically takes place over three stages: induction, consolidation, and maintenance.[3] Treatment is expected to span over approximately two years with the maintenance phase lasting the longest. T-ALL can spread to areas of the brain and spinal cord,[2] which can be diagnosed through lumbar puncture assessment in patients suspected to suffer from T-ALL. Lumbar puncture helps to identify leukemic cells surrounding the cerebrospinal fluid (CSF).[3] Even if leukemic cells are not found in the CSF at the time of diagnosis, it is highly likely that they will spread there with time and progression of the disease. Henceforth, prophylactic intrathecal chemotherapy in CNS lymphoma, a treatment to lower risk of leukemia spreading to the spinal cord and brain by directly administering chemotherapy to the CSF, is crucial.[3]

In comparison to B-ALL, T-ALL patients present more high-risk features including tendency for earlier relapse, CNS involvement, and resistance to chemotherapy. In response, Prophylactic Intrathecal Chemotherapy is further enhanced with CNS radiation therapy.[1] In treating high-risk T-ALL patients, allogeneic hematopoietic stem cell transplantation has been deemed to produce highly successful and promising results. However, its consequence includes increased relapse, which reduces its curative potential. Patients undergoing transplantation must be continuously monitored for minimal residual disease (MRD), usually via qPCR analysis of T-cell receptor (TCR) genes to evaluate for fusion transcripts such as SIL-TAL1.[15] Mutation of TAL1 is frequently present in T-ALL patients, where SIL/TAL1 fusion gives rise to inappropriate TAL1 expression, in turn promoting T-cell leukemogenesis.[4] The analysis is critical to ensure that immediate intervention is taken during early stages of relapse.

Young T-ALL patients showed significant improvement through multimodal therapy, involving initial induction therapy – including a glucocorticoid, vincristine, L-asparaginase, and an anthracycline – for 4 to 6 weeks, intensive combination therapy for 6–8 months, lastly 18–30 months of low-intensity anti-metabolite-based therapy.[8] It is crucial to note the importance of differential treatment amongst youth and adults. Studies have shown that through administering a random variation of either traditional pediatric scheme or intensive block-based chemotherapy, the two groups showed significantly different responses.[4] Although both treatments included administering high-dose methotrexate and asparaginase and allogeneic hematopoietic stem cell transplantation, high survival and low death rates were present for all patients for the first treatment whereas the latter led to a high toxic death rate amongst adults.[16]

Prognosis edit

In childhood, T-ALL patients can expect a 5-year event free survival and overall survival of, respectively, 70% and 80%.[1] Amongst approximately 25% of children who relapse, survival rate sits at 30-50% and the patients show much poorer prognosis.[1] Monitoring for MRD is critical as previously mentioned, through qPCR analysis, in order to evaluate the efficacy of treatment.

Recent genomic studies have found that a selection of genetic variants in relation to clonal evolution that drive resistance have been found as the basis for T-ALL relapse. Over 20% of patients with relapsed T-ALL showed mutation in the variant cytosolic 5’-nucleotidase II (NT5C2) gene, while the TFDP3 gene has also been found to confer chemoresistance in children.[1]

Epidemiology edit

Although over 100 genes mutations have been identified in T-ALL patients, only NOTCH1 and CDKN2A mutations are considered to be common.[13]

In over 50% of pediatric T-ALL cases, mutations in epigenetic regulators have been identified.[5] This activates mutations of NOTCH1 and FBXW7 causes the tumor-suppressing gene to lose its functions, leading to T-ALL.[17]

Near-telomeric location may sometimes generate subtle exchanges in DNA material at the loci involved in oncogenic rearrangements of T-ALL. This causes cryptic translocation and therefore deletes the putative tumor suppressor gene CDKN2A (INK4A). At the same time, TLX1 and NOTCH1 may also be activated at higher frequency than usual. The multistep prognosis of T-ALL has hence been said to intensify and rapidly progress due to accumulation of effects resulting from dysregulation of multiple signaling pathways.[5]

References edit

  1. ^ a b c d e f g h i j k l m n o "Pediatric T-Cell Acute Lymphoblastic Leukemia". atlasgeneticsoncology.org. Retrieved 2020-04-07.
  2. ^ a b c d e "Acute lymphoblastic leukemia (ALL): Symptoms, causes, and treatment". www.medicalnewstoday.com. 14 October 2019. Retrieved 2020-04-07.
  3. ^ a b c d "Typical Treatment of Acute Lymphocytic Leukemia (ALL)". www.cancer.org. Retrieved 2020-04-07.
  4. ^ a b c d D’Angiò, Mariella; Valsecchi, Maria G.; Testi, Anna M.; Conter, Valentino; Nunes, Vittorio; Parasole, Rosanna; Colombini, Antonella; Santoro, Nicola; Varotto, Stefania; Caniglia, Maurizio; Silvestri, Daniela (January 2015). "Clinical features and outcome of SIL/TAL1-positive T-cell acute lymphoblastic leukemia in children and adolescents: a 10-year experience of the AIEOP group". Haematologica. 100 (1): e10–e13. doi:10.3324/haematol.2014.112151. ISSN 0390-6078. PMC 4281327. PMID 25304610.
  5. ^ a b c "T-lineage acute lymphoblastic leukemia (T-ALL)". atlasgeneticsoncology.org. Retrieved 2020-04-07.
  6. ^ a b c d e Litzow, Mark R.; Ferrando, Adolfo A. (2015-08-13). "How I treat T-cell acute lymphoblastic leukemia in adults". Blood. 126 (7): 833–841. doi:10.1182/blood-2014-10-551895. ISSN 0006-4971. PMID 25966987.
  7. ^ "Acute Lymphoblastic Leukemia (ALL)". www.stjude.org. Retrieved 2020-04-07.
  8. ^ a b c d "T-cell Acute Lymphoblastic Leukaemia". Leukaemia Care. Retrieved 2020-04-07.
  9. ^ "What is the Difference Between B-cell Lymphoma and T-cell Lymphoma?". Dana-Farber Cancer Institute. 18 Jun 2019. Retrieved 7 Apr 2020.
  10. ^ a b c "T-Cell Acute Lymphoblastic Leukemia - My Cancer Genome". www.mycancergenome.org. Retrieved 2020-04-07.
  11. ^ a b "Leukemia - Chronic T-Cell Lymphocytic - Stages". Cancer.Net. 2012-06-25. Retrieved 2020-04-07.
  12. ^ Khalade, Abdul; Jaakkola, Maritta S.; Pukkala, Eero; Jaakkola, Jouni J. K. (2010-06-28). "Exposure to benzene at work and the risk of leukemia: a systematic review and meta-analysis". Environmental Health: A Global Access Science Source. 9: 31. doi:10.1186/1476-069X-9-31. ISSN 1476-069X. PMC 2903550. PMID 20584305.
  13. ^ a b "Human T-cell leukemia virus type 1 | Genetic and Rare Diseases Information Center (GARD) – an NCATS Program". rarediseases.info.nih.gov. Retrieved 2020-04-07.
  14. ^ a b "Acute lymphocytic leukemia - Diagnosis and treatment - Mayo Clinic". www.mayoclinic.org. Retrieved 2020-04-07.
  15. ^ D’Angiò, Mariella; Valsecchi, Maria G.; Testi, Anna M.; Conter, Valentino; Nunes, Vittorio; Parasole, Rosanna; Colombini, Antonella; Santoro, Nicola; Varotto, Stefania; Caniglia, Maurizio; Silvestri, Daniela (2015-01-16). "Clinical features and outcome of SIL/TAL1-positive T-cell acute lymphoblastic leukemia in children and adolescents: a 10-year experience of the AIEOP group". Haematologica. 100 (1): e10–e13. doi:10.3324/haematol.2014.112151. ISSN 0390-6078. PMC 4281327. PMID 25304610.
  16. ^ Quist-Paulsen, P.; Toft, N.; Heyman, M.; Abrahamsson, J.; Griškevičius, L.; Hallböök, H.; Jónsson, Ó G.; Palk, K.; Vaitkeviciene, G.; Vettenranta, K.; Åsberg, A. (2020-02-20). "T-cell acute lymphoblastic leukemia in patients 1–45 years treated with the pediatric NOPHO ALL2008 protocol". Leukemia. 34 (2): 347–357. doi:10.1038/s41375-019-0598-2. ISSN 1476-5551. PMID 31611626. S2CID 204459614.
  17. ^ Belver, Laura; Ferrando, Adolfo (2016-08-30). "The genetics and mechanisms of T cell acute lymphoblastic leukaemia". Nature Reviews Cancer. 16 (8): 494–507. doi:10.1038/nrc.2016.63. ISSN 1474-1768. PMID 27451956. S2CID 28636912. Retrieved 2020-04-05.

cell, acute, lymphoblastic, leukemia, confused, with, lymphoblastic, leukemia, lymphoma, adult, cell, leukemia, lymphoma, type, acute, lymphoblastic, leukemia, with, aggressive, malignant, neoplasm, bone, marrow, acute, lymphoblastic, leukemia, condition, wher. Not to be confused with T lymphoblastic leukemia lymphoma or Adult T cell leukemia lymphoma T cell acute lymphoblastic leukemia T ALL is a type of acute lymphoblastic leukemia with aggressive malignant neoplasm of the bone marrow 6 Acute lymphoblastic leukemia ALL is a condition where immature white blood cells accumulate in the bone marrow subsequently crowding out normal white blood cells 7 and create build up in the liver spleen and lymph nodes 8 The two most common types of ALL are B lymphocytes and T lymphocytes where the first protects the body against viruses and bacteria through antibody production which can directly destroy target cells or trigger others to do so whilst the latter directly destroy bacteria or cells infected with viruses 9 Approximately 20 of all ALL patients are categorized specifically to suffer from T ALL and it is seen to be more prevalent in the adult population in comparison to children with incidences spelling shown to diminish with age 6 10 Amongst T ALL cases in the pediatric population a median onset of age 9 has been identified and the disease is particularly prominent amongst adolescents 6 The disease stems from cytogenic and molecular abnormalities resulting in disruption of developmental pathways controlling thymocyte development tumor suppressor development and alterations in control of cell growth and proliferation 1 Distinct from adult T cell leukemia where T cell lymphotropic virus Type I causes malignant maturation of T cells T ALL is a precursor for lymphoid neoplasm 6 Its clinical presentation most commonly includes infiltration of the central nervous system CNS and further identifies mediastinal mass presence originating from the thymus along with extramedullary involvement of multiple organs including the lymph node as a result of hyperleukocytosis T cell acute lymphoblastic leukemiaT lymphoblastic cells of acute leukemia in the bone marrow In some cases the cytoplasm is concentrated at one pole of the cell forming hand mirror cells SpecialtyHaematology oncologySymptomsRecurrent infections unusual or common bleeding and bruising extreme tiredness unexplained fever unexplained weight gain swollen lymph nodesUsual onsetMost prevalent in the adult population with incidences spelling diminishing with age Amongst pediatric population median onset of age 9 Marked male predominance 1 CausesCurrently unknownDiagnostic methodBlood test bone marrow aspiration 2 biopsy CT MRI lumbar puncture 2 genetic testingTreatmentLong term chemotherapy 3 CNS radiation therapy 1 stem cell transplantation 4 Prognosis5 Year Event Free Survival 70 Overall Survival 80 1 Frequency7 at ages 1 10 14 at ages 10 15 and 29 at ages 15 18 5 Contents 1 Signs and symptoms 1 1 Clinical manifestations 2 Risk factors 3 Cytogenetics 4 Pathology 4 1 Causes of T ALL 4 1 1 Genetic conditions 4 1 2 Radiation exposure 4 1 3 Chemical exposure 4 1 4 Viruses 5 Diagnosis 5 1 Assessments 5 1 1 Blood tests 5 1 2 Bone marrow aspiration and biopsy 5 1 3 X rays and ultrasound 5 1 4 Lumbar puncture 5 1 5 Genetic test 5 2 Staging 6 Treatment 7 Prognosis 8 Epidemiology 9 ReferencesSigns and symptoms editT ALL patients may not always experience all the signs and symptoms below Patients with other medical conditions that are not leukemia may also experience similar symptoms Recurrent infections due to lack of normal white blood cells neutrophils 11 Unusual and or common bleeding and bruising Extreme tiredness and swellings in the neck lymph nodes or the middle of the chest causing possible facial swelling Unexplained fevers chills and or night sweats Unexplained weight loss and or loss of appetite Swollen lymph nodes Unexplained skin itchClinical manifestations edit Originating from epigenetic and genetic alterations in immature thymocytes T ALL is a highly aggressive and heterogenous disease Patients often present extensive bone marrow involvement mediastinal mass adenopathy CNS involvement and splenomegaly 1 Symptoms can be presented acutely or develop progressively over time The most common clinical feature amongst patients is the proliferation of malignant clones hence suppressing normal hematopoiesis resulting in deficiency of functioning peripheral blood cells particularly thrombocytes deficiency 1 Risk factors editT ALL is not a contagious nor inherited condition Its two main risk factors are age and gender 8 Most cases of leukemia increase with age with ALL being the main exception which peaks in children aged 2 to 5 years T ALL is seen to be most prevalent in the adult population but amongst cases in the pediatric population it is seen to have a median onset of age 9 and is most prominent to adolescents 6 10 The disease also is marked male predominance with a three fold increased risk of developing T ALL in comparison to females It is currently unclear as to why T ALL is preferential towards older children and males 1 Cytogenetics editBasic karyotyping showed structural chromosomal rearrangements in 50 75 of T ALL patients primarily inversion and translocations 1 Diagnostic yield can be substantially increased through further diagnosis through fluorescent in situ hybridization FISH and other various molecular technologies for example single nucleotide polymorphism SNP array The most common structural abnormality is rearrangement of the TCR gene 95 of T cell TCR consist of an alpha and beta chain encoded by TRA and TRB respectively where only 5 of T cell TCR consists of gamma and delta chains encoded by TRG and TRD respectively 4 Karyotyping showed that TRD and TRB undergo recombination most commonly whereas TRA is seldom involved and TRG is rarely rearranged These rearrangements affect the normal process of TCR and could lead to cellular machinery failing to correctly repair recombination activating RAG protein induced double strand breaks 1 All 30 genes known to illegitimately recombine with TCR genes function primarily to regulate epigenetics through roles such as signal transducers transcription factors tumor suppressors or oncogenes cell cycle regulators or ribosomal proteins T Cell TCR encoded by TRA TRD and TRG at chromosome bands 14q11 and 7q34 become malignant in T ALL patients 1 The build up of malignant T cells in T ALL are clones with identical T cell receptor gene arrangements having taken rise from a single cell The gene rearrangements as a result of the malignant cell juxtapose both TCR genes and other critical genes that code for transcription factors This results in dysregulation of partner gene transcription which serves as the main cause of leukemogenesis a multi step process of induction development and progression of leukemic diseases 1 20 of all leukemias demonstrate simultaneous rearrangement of these genes Pathology editLike most cancers mutations in the DNA begin T ALL development and lead to loss of function of white blood cells Different subtypes of leukemia have similarities in their causes which are a combination of genetics epigenetic changes and environmental factors However because there are few T ALL cases in comparison to other subtypes of leukemia there is currently no clear cause of T ALL T ALL is not contagious nor inherited but specific genetic mutations commonly including NOTCH1 and CDKN2A may be passed along which increases susceptibility of T ALL 10 Causes of T ALL edit Genetic conditions edit Some patients may have familial histories with leukemia predispositions which increases risk of developing T ALL Li Fraumeni syndrome is an inherited condition that leads to mutation of TP53 a tumor suppressor gene which then increases risk of T ALL Mutation in gene SPRED1 is also associated with development of T ALL Patients with immature thymocytes in the thymus begins T ALL development Furthermore hereditary conditions such as Down syndrome neurofibromatosis type 1 ataxia telangiectasia and Noonan syndrome are associated with higher risk of developing T ALL Radiation exposure edit source source source source Human T Lymphotropic VirusThose who have had previous chemotherapy and exposure to radiation may have increased risks of developing T ALL CDKN2A is an inherited polymorphism variant that is seen to be associated with development of T ALL SR 90 emission from nuclear reactor accidents is also believed to increase risk of developing T ALL Chemical exposure edit Benzene a chemical classified as being carcinogenic to humans is associated with increased risk of T ALL as well as other forms of leukemia 12 Viruses edit Human T lymphotropic virus HTLV 1 is a retroviral infection that affect white blood cells T cells which may later develop into T ALL and other subtypes of leukemia 13 Diagnosis editWhen doctors are suspicious of a patient potentially suffering from T ALL after careful examination of background including medical history signs and symptoms doctors would then conduct tests procedures and scans to proceed with diagnosis of T ALL Some symptoms and medical history may not be specific enough to diagnose T ALL so further testing may be required Doctors may consider some factors mentioned but would not necessarily conduct all tests possible 11 Assessments edit Blood tests edit Complete blood count CBC is done to test for T ALL by measuring the different types and maturity of cells in the patient s blood which allows the donor to determine whether leukemic cells are present in the patient Additionally blood tests that show high levels of white blood cells or low levels of red blood cells may also be a sign of T ALL Further testing could also help indicate whether T ALL has affected other organs such as the kidneys as well as the genetic alterations of the disease Bone marrow aspiration and biopsy edit Bone marrow consists of a combination of solid and liquid components Bone marrow aspiration and biopsies are typically done simultaneously to help determine and confirm the type and severity of T ALL Further biopsies such as skin and lymph node biopsies may also need to be done to check for the spread of T ALL 2 X rays and ultrasound edit As swollen spleen and lymph nodes are symptoms of T ALL X rays and ultrasound scans such as CT and MRI can help confirm the diagnosis This also provides information on the impact T ALL has on other organs of the body Lumbar puncture editTo prevent ineffective treatments towards T cells that have invaded the CNS lumbar puncture allows doctors to determine whether the treatments will be effective This also reveals the spread of T ALL 2 nbsp Lumbar Puncture PositionsGenetic test edit Genetic testing helps identify chromosomal abnormalities in the patients This can help identify the genetic mutations and therefore diagnose the specific leukemia subtype Staging edit Normal staging is not used for T ALL because it is already spread throughout the body when first diagnosed However they have their own system of classifying T ALL cases 14 First patterns of gene expression are investigated to define T ALL Then stages of thymic development can be determined by identifying specific expressions in chromosomal abnormalities This forms the stages of T ALL cases being either at high or low risk 8 Patients will then receive the appropriate treatment in respect to whichever class they are in 14 Treatment editCurrently standard treatment of T ALL takes the form of long term chemotherapy and drug intake to prevent or treat side effects associated with low white blood cell count as a result of intensive chemotherapy regimes The treatment typically takes place over three stages induction consolidation and maintenance 3 Treatment is expected to span over approximately two years with the maintenance phase lasting the longest T ALL can spread to areas of the brain and spinal cord 2 which can be diagnosed through lumbar puncture assessment in patients suspected to suffer from T ALL Lumbar puncture helps to identify leukemic cells surrounding the cerebrospinal fluid CSF 3 Even if leukemic cells are not found in the CSF at the time of diagnosis it is highly likely that they will spread there with time and progression of the disease Henceforth prophylactic intrathecal chemotherapy in CNS lymphoma a treatment to lower risk of leukemia spreading to the spinal cord and brain by directly administering chemotherapy to the CSF is crucial 3 In comparison to B ALL T ALL patients present more high risk features including tendency for earlier relapse CNS involvement and resistance to chemotherapy In response Prophylactic Intrathecal Chemotherapy is further enhanced with CNS radiation therapy 1 In treating high risk T ALL patients allogeneic hematopoietic stem cell transplantation has been deemed to produce highly successful and promising results However its consequence includes increased relapse which reduces its curative potential Patients undergoing transplantation must be continuously monitored for minimal residual disease MRD usually via qPCR analysis of T cell receptor TCR genes to evaluate for fusion transcripts such as SIL TAL1 15 Mutation of TAL1 is frequently present in T ALL patients where SIL TAL1 fusion gives rise to inappropriate TAL1 expression in turn promoting T cell leukemogenesis 4 The analysis is critical to ensure that immediate intervention is taken during early stages of relapse Young T ALL patients showed significant improvement through multimodal therapy involving initial induction therapy including a glucocorticoid vincristine L asparaginase and an anthracycline for 4 to 6 weeks intensive combination therapy for 6 8 months lastly 18 30 months of low intensity anti metabolite based therapy 8 It is crucial to note the importance of differential treatment amongst youth and adults Studies have shown that through administering a random variation of either traditional pediatric scheme or intensive block based chemotherapy the two groups showed significantly different responses 4 Although both treatments included administering high dose methotrexate and asparaginase and allogeneic hematopoietic stem cell transplantation high survival and low death rates were present for all patients for the first treatment whereas the latter led to a high toxic death rate amongst adults 16 Prognosis editIn childhood T ALL patients can expect a 5 year event free survival and overall survival of respectively 70 and 80 1 Amongst approximately 25 of children who relapse survival rate sits at 30 50 and the patients show much poorer prognosis 1 Monitoring for MRD is critical as previously mentioned through qPCR analysis in order to evaluate the efficacy of treatment Recent genomic studies have found that a selection of genetic variants in relation to clonal evolution that drive resistance have been found as the basis for T ALL relapse Over 20 of patients with relapsed T ALL showed mutation in the variant cytosolic 5 nucleotidase II NT5C2 gene while the TFDP3 gene has also been found to confer chemoresistance in children 1 Epidemiology editAlthough over 100 genes mutations have been identified in T ALL patients only NOTCH1 and CDKN2A mutations are considered to be common 13 In over 50 of pediatric T ALL cases mutations in epigenetic regulators have been identified 5 This activates mutations of NOTCH1 and FBXW7 causes the tumor suppressing gene to lose its functions leading to T ALL 17 Near telomeric location may sometimes generate subtle exchanges in DNA material at the loci involved in oncogenic rearrangements of T ALL This causes cryptic translocation and therefore deletes the putative tumor suppressor gene CDKN2A INK4A At the same time TLX1 and NOTCH1 may also be activated at higher frequency than usual The multistep prognosis of T ALL has hence been said to intensify and rapidly progress due to accumulation of effects resulting from dysregulation of multiple signaling pathways 5 References edit a b c d e f g h i j k l m n o Pediatric T Cell Acute Lymphoblastic Leukemia atlasgeneticsoncology org Retrieved 2020 04 07 a b c d e Acute lymphoblastic leukemia ALL Symptoms causes and treatment www medicalnewstoday com 14 October 2019 Retrieved 2020 04 07 a b c d Typical Treatment of Acute Lymphocytic Leukemia ALL www cancer org Retrieved 2020 04 07 a b c d D Angio Mariella Valsecchi Maria G Testi Anna M Conter Valentino Nunes Vittorio Parasole Rosanna Colombini Antonella Santoro Nicola Varotto Stefania Caniglia Maurizio Silvestri Daniela January 2015 Clinical features and outcome of SIL TAL1 positive T cell acute lymphoblastic leukemia in children and adolescents a 10 year experience of the AIEOP group Haematologica 100 1 e10 e13 doi 10 3324 haematol 2014 112151 ISSN 0390 6078 PMC 4281327 PMID 25304610 a b c T lineage acute lymphoblastic leukemia T ALL atlasgeneticsoncology org Retrieved 2020 04 07 a b c d e Litzow Mark R Ferrando Adolfo A 2015 08 13 How I treat T cell acute lymphoblastic leukemia in adults Blood 126 7 833 841 doi 10 1182 blood 2014 10 551895 ISSN 0006 4971 PMID 25966987 Acute Lymphoblastic Leukemia ALL www stjude org Retrieved 2020 04 07 a b c d T cell Acute Lymphoblastic Leukaemia Leukaemia Care Retrieved 2020 04 07 What is the Difference Between B cell Lymphoma and T cell Lymphoma Dana Farber Cancer Institute 18 Jun 2019 Retrieved 7 Apr 2020 a b c T Cell Acute Lymphoblastic Leukemia My Cancer Genome www mycancergenome org Retrieved 2020 04 07 a b Leukemia Chronic T Cell Lymphocytic Stages Cancer Net 2012 06 25 Retrieved 2020 04 07 Khalade Abdul Jaakkola Maritta S Pukkala Eero Jaakkola Jouni J K 2010 06 28 Exposure to benzene at work and the risk of leukemia a systematic review and meta analysis Environmental Health A Global Access Science Source 9 31 doi 10 1186 1476 069X 9 31 ISSN 1476 069X PMC 2903550 PMID 20584305 a b Human T cell leukemia virus type 1 Genetic and Rare Diseases Information Center GARD an NCATS Program rarediseases info nih gov Retrieved 2020 04 07 a b Acute lymphocytic leukemia Diagnosis and treatment Mayo Clinic www mayoclinic org Retrieved 2020 04 07 D Angio Mariella Valsecchi Maria G Testi Anna M Conter Valentino Nunes Vittorio Parasole Rosanna Colombini Antonella Santoro Nicola Varotto Stefania Caniglia Maurizio Silvestri Daniela 2015 01 16 Clinical features and outcome of SIL TAL1 positive T cell acute lymphoblastic leukemia in children and adolescents a 10 year experience of the AIEOP group Haematologica 100 1 e10 e13 doi 10 3324 haematol 2014 112151 ISSN 0390 6078 PMC 4281327 PMID 25304610 Quist Paulsen P Toft N Heyman M Abrahamsson J Griskevicius L Hallbook H Jonsson o G Palk K Vaitkeviciene G Vettenranta K Asberg A 2020 02 20 T cell acute lymphoblastic leukemia in patients 1 45 years treated with the pediatric NOPHO ALL2008 protocol Leukemia 34 2 347 357 doi 10 1038 s41375 019 0598 2 ISSN 1476 5551 PMID 31611626 S2CID 204459614 Belver Laura Ferrando Adolfo 2016 08 30 The genetics and mechanisms of T cell acute lymphoblastic leukaemia Nature Reviews Cancer 16 8 494 507 doi 10 1038 nrc 2016 63 ISSN 1474 1768 PMID 27451956 S2CID 28636912 Retrieved 2020 04 05 Retrieved from https en wikipedia org w index php title T cell acute lymphoblastic leukemia amp oldid 1144480813, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.