Friday 18 December 2015

THERAPEUTIC GOAL OF HYPOPLASTIC ANEMIA IN PEDIATRICS HOSPITAL IN INDIA

By Health Basic   Posted at  09:18   No comments

Aleem sarwar,
Pharm.d. Interns.
Dept.of pharmacy practice,
Annamalai University,
Chidambaram,608 002,
Tamil Nadu,India
mob.9962761548.

 
 Abstract
 Aplastic anemia is acondition in which the bone marrow does not make enough new blood cells. bone marrow is the soft, fatty tissue in the center of  bones.
Most acquired aplastic anemia (AA) is the result of immune-mediated
destruction of hematopoietic stem cells causing pancytopenia and an empty bone marrow, which
can be successfully treated with either immunosuppressive therapy (IST) or hematopoietic stem-
cell transplantation (HSCT).the aplastic anemia is a rare and serious disease.It happens when your bone marrow fails to make enough blood cells (low blood cell counts).

Keywords
  aplastic anemia; anti-thymocyte globulin; bone marrow failure; stem cell transplantation;
cyclosporine; pancytopenia

Itroduction
 Aplastic anemia (AA) is a life-threatening bone marrow failure disorder
which, if untreated, is associated with very high mortality. Hematopoietic stem cell
transplantation (HSCT) offers an opportunity for cure, but most patients are not suitable
candidates for this procedure due to advanced age, comorbidities, or lack of a
histocompatible donor. For these patients, comparable long term survival is attainable with
immunosuppressive treatment (IST) with anti-thymocyte globulin (ATG) and cyclosporine
(CsA). Although several etiopathogenic triggers have been proposed in AA, the majority of
cases are idiopathic, with a small percentage of cases occurring after an episode of
seronegative hepatitis.
 Epidemiology and etiology
Each year, approximately between  800 to 1200 indian learn that they have aplastic anemia.the disease can strike people of any age, race, and gender. But it more common among children, teenagers, and young adults.

About 1 in 4 cases aplastic anemia can be linked to one of sveral causes. 
These include.
  • Toxins, such as pesticides, arsenic, and benzene.
  • Treatments for other autoimmune disease such as rheumatoid arthritis.
  • Pregnancy. some times this aplastic anemia improves on its own after the woman give birth.                                                                                                                                            AA was attributed to an idiosyncratic reaction to drug or chemical
    exposure. The association of medical drug use to AA is of great importance, as it is
    devastating to patients and physicians and presents serious legal consequences and problems
    in pharmaceutical drug development. However, the study of idiosyncratic drug reactions,
    by definition extremely rare, is difficult and the only clear predisposition to abnormal drug
    metabolism underlying susceptibility is one study of a single individual exposed to
    carbamazepine published over 20 years ago. Overrepresentation of drug metabolizing
    glutathione-S-transferase gene deletions have been observed in some series  but no
    reasonable mechanism has been developed for chloramphenicol.
    Pregnancy and eosinophilic fasciitis are linked to AA. Five to ten percent of cases of AA
    follow an episode of seronegative hepatitis, but despite intensive efforts, an infectious
    agent has not been identified.
Pathophysiology                                                                                                                                     
 In most cases, AA behaves as an immune-mediated disease. An immune response
dominated by oligoclonal expanded cytotoxic T-cells targets hematopoietic stem and
progenitor cells, inducing their death via apoptosis and hematopoietic failure.
T-Cell-mediated destruction of the bone marrow
Recovery of autologous hematopoiesis in patients who failed to engraft after stem cell
transplant and responsiveness to immunosuppressive therapies are the major clinical
evidences supporting an immune pathophysiology underlying acquired AA. Although a
nonimmune pathophysiology has been inferred from a failure to respond to
immunosuppression, refractoriness to therapy is also consistent with very severe stem cell
depletion, a “spent” immune response, or immunological mechanisms resistant to current
therapies.
Removal of lymphocytes from aplastic bone marrows improves colony numbers in tissue
culture, and their addition to normal marrow inhibited hematopoiesis in vitro. The
effector cells within the lymphocyte subset are activated cytotoxic T cells bearing a
profile, expressing and secreting interferon-γ. 
T-bet, a transcription factor that binds tothe interferon-γ
promoter region and is critical for Th1 polarization, is up-regulated in T-
cells of patients with AA. Specific CD8+CD28− cell clones are expanded in AA
peripheral blood, as manifest by skewed usage of the Vβ repertoire; and oligoclones
recognize and induce apoptosis of autologous myeloid cells. Regulatory T cells, which
control and suppress auto-reactive T cells, are decreased at presentation in almost all
patients with AA. In a mouse model of immune-mediated marrow failure, addition of T
regulatory cells abrogated pancytopenia induced by the infusion of lymph node cells.
Why T-cells are activated in AA is unclear. HLA-DR2 is over-represented among patients,
suggesting a role for antigen recognition, and its presence is predictive of a better response
to cyclosporine. Polymorphisms in cytokine genes, associated with an increased
immune response, also are more prevalent, such as for tumor necrosis factor-α(TNF2)
promoter at −308, interferon-γ, and interleukin 6 genes. These alterations in
nucleotide sequence and in gene regulation suggest a genetic basis for aberrant T cell
activation in bone marrow failure.
Hematopoiesis

Immune attack leads to marrow failure. The targets, mainly CD34+ cells, are very few or
absent in the aplastic bone marrow, and minimal numbers of colonies derive from
committed progenitors in semisolid media, all reflecting the severe reduction in
hematopoietic progenitor cells that defines the disease. The reduced number and function of
the marrow is secondary to cell destruction, and apoptosis is prevalent among the few
remaining elements. Microarray analysis of the remaining CD34+ cells shows a
transcriptome shifted towards apoptosis, cell death, and immune regulation.
A minority of AA cases may share pathophysiologic basis with inherited marrow failure
syndromes. One peculiar feature of white blood cells in AA is short telomeres, observed in
approximately a third of cases. Although initially blamed on excessive stem cell
turnover, telomere shortening in some cases of acquired AA and in dyskeratosis congenita, a
constitutional marrow failure syndrome is due to mutations in components of the telomerase
complex, causing low telomerase activity, progressive telomere erosion, and a deficient
proliferative capacity of hematopoietic stem cells. Family members who share the
mutation, despite normal or near normal blood counts, have hypocellular marrows, reduced
CD34+ cell counts and poor hematopoietic colony formation, increased hematopoietic
growth factor levels, and of course short telomeres; some affected relatives may present with
pulmonary fibrosis or liver cirrhosis in isolation. A few adult AA patients also have
heterozygous mutations in the Shwachman-Bodian-Diamond syndrome (SBDS) gene
(Shwachman-Diamond syndrome occurs when patient carries biallelic mutations).
Clonal evaluation
AA may coexist or evolve to clonal disorders, as paroxysmal nocturnal hemoglobinuria
(PNH), myelodysplasia (MDS), or acute myeloid leukemia (AML).
Paroxysmal nocturnal hemoglobinuria (PNH)

About 40–50% of patients with acquired AA have expanded populations of PNH cells,
easily detected by flow cytometry due to the absence of glycosylphosphatidylinositol-linked
membrane proteins, the result of somatic PIG-A gene mutations arising in hematopoietic
stem cells. Most clones are small and do not lead to clinical manifestations of
hemolysis or thrombosis, but classic PNH can evolve to marrow failure (the AA/PNH
syndrome) and all PNH patients show evidence of underlying hematopoietic deficiency. The
global absence of large number of cell surface proteins in PNH has been hypothesized to
allow escape and survival of a pre-existing mutant clone.
Myelodysplasia (MDS)
Aneuploidy develops in a minority of patients treated with immunosuppression over time,
usually monosomy 7 and trisomy 8. AA patients who develop trisomy 8 usually respond
to IST. T-cell oligoclones appear to recognize the aneuploid cells and specifically WT1
 as an antigen, but target cells are not killed due to their expression of anti-apoptotic genes.
Immunosuppressive therapy
Horse anti-thymocyte globulin (ATGAM (R); h-ATG) is the only drug approved by the
Food and Drug Administration for the treatment of AA. While it is generally believed that h-ATG administration leads to depletion of immune competent cells, its exact mechanism of
action remains unclear. H-ATG preparations contain a variety of antibodies recognizing
human T-cell epitopes, many directed against activated T-cells or activation antigens.

Both refractory and relapsed patients are frequently treated with further courses of ATG. In
these settings, rabbit ATG (r-ATG) + CsA has been frequently used. R-ATG is similar to h-ATG except that gamma immune globulin is obtained by immunization of rabbits with
human thymocytes. Clinically, r-ATG appears to be more immunosuppressive as a more
prolonged lymphopenia is observed with this agent compared to h-ATG. This enhanced
lymphocytotoxicity of r-ATG may be explained by higher affinity IgG subtype to human
lymphocytes, less batch-to-batch variability, longer half-life, and more efficient lymphocyte
depletion. In addition, r-ATG may promote immune regulation as suggested by an in
vitro assay where CD4+CD25− were converted to CD4+CD25+ regulatory T cells in the
presence of r-ATG but not h-ATG. For the 1/3 of patients who are refractory to h-ATG/CsA, repeated courses of immunosuppression have yielded response rates varying from 30 –70%. For relapsed patients, re-introduction of CsA commonly result in
improvement in blood counts, however, CsA-dependence is frequent and the dose of CsA
usually is tapered slowly with hematologic monitoring. Re-treatment with ATG/CsA in
relapsed AA has resulted in response rates of 50–60%. In our experience, relapse
 does not correlate to a poor prognosis, as patients often respond to re-introduction of CsA and/or re-treatment with ATG
 In moderate AA, the clinical course is variable: some patients progress to severe disease,
others remain stable and may not require intervention; regular transfusions may not be
required. Very few clinical trials have specifically addressed moderate disease.
Immunosuppression can reverse moderate pancytopenia and alleviate transfusion
requirements; ATG and cyclosporine are more effective in combination. Daclizumab, a
humanized monoclonal antibody to the interleukin-2 receptor, improved blood counts and
relieved transfusion requirements in 6 of 16 evaluable patients; the outpatient regimen had
little toxicity.
Allogeneic Hematopoietic Stem Cell Transplantation
Allogeneic bone marrow transplantation from a histocompatible matched sibling is curative
therapy in the majority of SAA patients who undergo this procedure . The most recent
cohort reported to the IBMTR showed 77% 5-year survival, and in children and patients
who were minimally transfused, survival of 80–90% may be routinely achieved. Acute
grade II–IV graft-versus-host-disease (GVHD) occurs in about 20–30% of patients and
chronic GVHD in 30–40%. Chronic GVHD has been a major cause of morbidity
and mortality in patients who survived more than 2 years post graft, with the necessity of
long term immunosuppressive therapy common. Graft rejection, a historic problem in
the application of transplant to SAA is now infrequent, likely a benefit of less immunogenic
blood products (leukocyte-depleted erythrocytes, for example) from fewer donors (platelets
collected by cytoapheresis). Also, better radiation-free conditioning regimens have
 improved the tolerability of HSCT and allowed for engraftment in heavily transfused (and in
some cases alloimmunized) patients who were refractory to IST. The frequently
employed conditioning regimen of cyclophosphamide + ATG was compared to
cyclophosphamide in a prospective randomized study  no differences in engraftment,
GVHD, and survival rates were observed between the two groups, suggesting that the better
outcome with HSCT over time relates to advances in supportive care. The main source of
stem cells up to 2000 has been bone marrow cells; in recent years, G-CSF peripheral blood
(PB) mobilized CD34+ have become more widely used. In a retrospective analysis, the use
of PB progenitor cell graft has been correlated to a worse outcome and more chronic GVHD
in younger patients (less than 20) compared to those who receive a bone marrow graft in
HLA-matched sibling donor transplants. This difference was not observed in older
patients.
Matched unrelated donor transplant
A matched sibling donor is available in only 20–30% of cases. As the outcome in aplastic
patients who have failed a single round of ATG has been poor, alternative sources of
hematopoietic stem cells have been sought, usually from now very large donor registries.
Data from large retrospective studies suggest that the outcome for an unrelated donor HSCT
remains less favorable compared to a matched-related transplant, due to more GVHD, a
mortality rate that is about twice that observed in matched sibling transplants, and long term
survival of about 50%  Older patients with poor performance status have the
worst outcome and better results are obtained in children than for adults. Progress in donor
selection through high-resolution HLA typing technology has likely contributed to
decreased graft rejection and better survival  and recently reported outcomes for MUD
rival those for an HLA-matched sibling transplant in children and young adults.
Conclusion
In recent years, further evidence has accumulated to strengthen the hypothesis that bone
marrow failure in AA results from immunologic destruction of hematopoietic stem and
progenitor cells. In a minority of patients with shortened telomeres, a qualitative disorder
may accompany the numerical diminution of CD34+ cells in the bone marrow. Current
research is aimed at investigating the mechanisms that lead to T cell activation and, whether
it is antigen driven or a result of immunological disarray. The clinical implication of
telomere shortening in AA is being investigated for prognostic significance. Most patients
with AA are now expected to survive regardless of the treatment modality employed.
Advances in supportive care and better salvage therapies have produced significantly
increased survival among initial non-responders to ATG+ CsA; the decrease in allo-
sensitization (through routine leucodepletion of blood products) and better donor selection
(through high molecular HLA-typing) have improved the outcomes of both related and
unrelated HSCT; long term survival in pediatric patients who respond to IST is excellent and
 ATG based regimen should be offered as initial therapy to those who lack a matched sibling
donor. Our practice has been to consider an alternative donor HSCT in suitable pediatric
patients who have failed an initial course of ATG + CSA or in adults who have failed two
courses of IST. Refractory patients who are not suitable candidates for alternative donor
HSCT can be supported through a combination of transfusions, androgens and/or
administration of hematopoietic growth factors. In chronically transfused patients, iron
overload can now be managed with more convenient oral agents (deferasirox, Exjade) and,
the prospect of approval of newer oral thrombopoietic drugs is likely to ease platelet
transfusion requirements especially in AA patients who are refractory to IST.
ATG based regimen should be offered as initial therapy to those who lack a matched sibling
donor. Our practice has been to consider an alternative donor HSCT in suitable pediatric
patients who have failed an initial course of ATG + CSA or in adults who have failed two
courses of IST. Refractory patients who are not suitable candidates for alternative donor
HSCT can be supported through a combination of transfusions, androgens and/or
administration of hematopoietic growth factors. In chronically transfused patients, iron
overload can now be managed with more convenient oral agents (deferasirox, Exjade) and,
the prospect of approval of newer oral thrombopoietic drugs is likely to ease platelet
transfusion requirements especially in AA patients who are refractory to IST.
References

1. Kaufman, DW.; Kelly, JP.; Levy, M.; Shapiro, S. The Drug Etiology of Agranulocytosis and
Aplastic Anemia. Oxford; New York: 1991.
*2. Issaragrisil S, Kaufman D, Anderson T, Chansung K, Leaverton P, Shapiro S, Young NS, The
Aplastic Anemia Study G. The epidemiology of aplastic anemia in Thailand. Blood. 2006;
107:1299–1307. [PubMed: 16254144] This large epidemiologic study enrolled more than 500
patients and 2,000 case controls; an accurate incidence rate was obtained, and there were
environmental associations with the disease that were familiar (chemicals and drugs) and others
that were unexpected (water source, animal exposure).
3. Hamerschlak N, Maluf E, Pasquini R, Eluf-Neto J, Moreira FR, Cavalcanti AB, Okano ÍR, Falcão
RP, Pita MT, Loggetto SR, et al. Incidence of aplastic anemia and agranulocytosis in Latin
America: the LATIN study. Sao Paulo Medical Journal. 2005; 123:101–104. [PubMed: 16021270]
4. Young, NS.; Young, NS.; Alter, BP. Drugs and chemicals. In: Saunders, WB., editor. Aplastic
Anemia, Acquired and Inherited. 1994. p. 100-132.
5. Gerson WT, Fine DG, Spielberg SP, Sensenbrenner LL. Anticonvulsant-induced aplastic anemia:
increased susceptibility to toxic drug metabolites in vitro. Blood. 1983; 61:889–893. [PubMed:
6831050]
6. Sutton JF, Stacey M, Kearsey SE, Reig TS, Young NS, Liu JM. Increased risk for aplastic anemia
and myelodysplastic syndrome in individuals lacking GSTT1 gene. Pediatric Blood Cancer. 2004;
42:122–126. [PubMed: 14752874]
7. Dufour C, Syahn J, Bacigalupo A, Longoni D, Varotto S, Iori AP, Bagnasco F, Locasciulli A,
Menna G, Ramenghi U, et al. Genetic polymorphisms of CYP3A4, GSTT1, GSTM1, GSTP1 and
NQO1 and the risk of acquired idiopathic aplastic anemia in Caucasian patients. Haematologica.
2005; 90:1027–1031. [PubMed: 16079101]
8. Lu J, Basu A, Melenhorst J, Young NS, Brown KE. Analysis of T-cell repertoire in hepatitis-
associated aplastic anemia. Blood. 2004; 103:4588–4593. [PubMed: 14988156]
9. Young NS. Hematopoietic cell destruction by immune mechanisms in aquired aplastic anemia.
Seminars in Hematology. 2000; 37:3–14. [PubMed: 10676907]
10. Sloand EM, Kim S, Maciejewski JP, Tisdale J, Follmann D, Young NS. Intracellular interferon- y
in circulating and marrow T cells detected by flow cytometry and the response to
imunosuppressive therapy in patients with aplastic anemia. Blood. 2002; 100:1185–1191.
[PubMed: 12149196]
*11. Solomou EE, Keyvanfar K, Young NS. T-bet, a Th1 transcription factor, is up-regulated in T cells
from patients with aplastic anemia. Blood. 2006; 107:3983–3991. [PubMed: 16434488] The T
cell polarizing transcription factor T-bet was increased in T cells from most patients with aplastic
anemia.
12. Risitano AM, Maciejewski JP, Green S, Plasilova M, Zeng W, Young NS. In-vivo dominant
immune responses in aplastic anaemia: molecular tracking of putatively pathogenetic T-cell clones
by TCR [beta]-CDR3 sequencing. The Lancet. 2004; 364:355–364.
**13. Solomou EE, Rezvani K, Mielke S, Malide D, Keyvanfar K, Visconte V, Kajigaya S, Barrett AJ,
Young NS. Deficient CD4+ CD25+ FOXP3+ T regulatory cells in acquired aplastic anemia.
Blood. 2007; 110:1603–1606. [PubMed: 17463169] Regulatory T cells were decreased at
presentation in almost all patients; a regulatory T cell defect 16 was implicated in autoimmune.
14. Aplastic anemia and MDS international foundation.

About the Author

Nulla sagittis convallis arcu. Sed sed nunc. Curabitur consequat. Quisque metus enim, venenatis fermentum, mollis in, porta et, nibh. Duis vulputate elit in elit. Mauris dictum libero id justo.
View all posts by: BT9

0 comments:

Back to top ↑
Connect with Us

What they says

© 2013 HEALTH BASIC. Distributed By Blogger Themes | WP Mythemeshop Converted by Bloggertheme9
Blogger templates. Proudly Powered by Blogger.