Skip to main content
Oncology Reviews logoLink to Oncology Reviews
. 2020 Jul 14;14(2):466. doi: 10.4081/oncol.2020.466

CD markers polymorphisms as prognostic biomarkers in hematological malignancies

Saeid Shahrabi 1, Majid Ghanavat 2, Masumeh Maleki Behzad 3,4, Daryush Purrahman 4, Najmaldin Saki 4,
PMCID: PMC7385526  PMID: 32782727

Abstract

The clusters of differentiation (CD) are surface molecules used for immunophenotyping of cells. The expression of CD markers is widely used to classify hematological malignancies, including leukemia and lymphoma. Single nucleotide polymorphisms (SNPs) are crucial genetic changes that can be associated with abnormal expression and function of CD markers. In this paper, we assess the prognostic effect of CD markers SNPs in hematological malignancies. Materials and methods and relevant literature was identified by a PubMed search (2001-2019) of English language papers using the following terms: ‘polymorphism’, ‘CD marker’, ‘leukemia’, ‘lymphoma’, ‘prognosis’, ‘CD marker’, and ‘polymorphism’. Many studies have demonstrated the effects of CD markers polymorphisms on risk of hematological malignancies. Also, SNPs of CD markers can be related with clinicopathological features, invasiveness, and response to therapy of these disorders. Considering the importance of SNPs in the expressions of CD markers, these genetic changes could be used as potential prognostic biomarkers in hematological malignancies. It is hoped that the evaluation of SNPs in CD markers will enable early diagnosis, prognosis, and detection of response to treatment. However, better understanding of SNPs in CD markers that are involved in hematological malignancies requires further studies on different populations of the worldwide.

Key words: Leukemia, lymphoma, CD marker, polymorphism, prognosis

Introduction

The cluster of differentiation (CD) markers are the key to distinguish and differentiate between cells. Surface CD markers have specific functions depending on the cell type and can be differently expressed in response to environmental conditions and intracellular genetic changes.1 Therefore, the detection of CD markers can be a useful approach to identify abnormal cells in disorders such as leukemia. Leukemia and lymphoma are a group of hematological malignancies originating from bone marrow (BM), which are related with high production and proliferation but non-differentiation of hematopoietic stem cells (HSCs). Accumulation of blast cells in BM, peripheral blood (PB), and lymph nodes leads to clinical manifestations among patients.2 Abnormal expression of CD markers in BM and PB of patients is one of the first diagnostic features of leukemia. Therefore, the evaluation of CD markers’ expressions is used as the first diagnostic strategy that is followed by an appropriate approach to monitoring the clinical course of leukemia.3 Despite the fact that specific immunophenotypic patterns have been defined for the diagnosis of leukemia and lymphoma, results of studies in recent years indicate the heterogeneous expressions of CD markers in these malignancies. Numerous environmental and genetic factors like single nucleotide polymorphisms (SNPs) can alter the expression patterns of these surface markers. Although cytogenetic alterations have been identified as an underlying cause of leukemia progression, SNPs can change the expression and function of CD markers and affect various aspects in the progression of these disorders, including the clinical course and response to treatment in patients.4 Therefore, detection of the primary mechanisms of changes in CD markers expressions in leukemia may contribute to the prognosis of these disorders.

Recent developments in molecular biology and the discovery of the association between SNPs of different genes with leukemia/lymphoma progression have highlighted the critical role of this genetic changes in the prognosis of these malignancies. Therefore, in this paper we discuss the most common SNPs in CD markers and their possible mechanisms for leukemia/lymphoma prognosis.

Effect of CD markers’ polymorphisms on the clinical outcome of hematological malignancies

CD33

The presence of cytogenetic aberrations at the time of diagnosis is a major prognostic factor for acute myeloid leukemia (AML). However, studies have indicated that SNPs are among the genetic changes that can be associated with the prognosis of this malignancy by altering the expression and function of CD markers in AML blasts. CD33 is a well-known marker on the surface of AML blasts, which is targeted by chemotherapeutic agents such as gemtuzumab ozagamicin (GO) as a routine treatment of AML.5 Mortland et al. in their study showed that rs2455069, rs1803254, and rs12459419 SNPs were associated with decreased CD33 expression on the surface of blasts in children with AML.4 Although the primary role of CD33 has not been elucidated, it seems to be involved in both proliferation and survival of AML blasts.6 Hence, it is inferred that SNPs related to decreasing CD33 expression are associated with good prognosis in children with AML. In addition, decreased expression of CD33 reduces the need for GO therapy and its accompanying complications for patients, including shivering, fever, nausea, and vomiting.7 Furthermore, researchers showed that the TT genotype derived from rs12459419 SNP that involves exon 2 of CD33 gene, as well as carrying at least one C allele of rs1803254 SNP leads to better overall survival (Table 1).4 Nevertheless, patients harboring AA or AG alleles caused by rs35112940 have a higher risk of recurrence and show decreased survival.4 In fact, CD33 SNPs appear to be a prognostic factor for assessing clinical course and response to treatment in AML patients through influencing clinical features.

CD44

CD44, which plays an important roles in cellular adhesion and migration, is a surface receptor on a large number of cells (including HSCs) that interacts with various ligands such as hyaluronic acid.8 Therefore, increasing CD44 expression could account for homing of normal HSCs or malignant cells in BM niches. Research has shown that CD44 rs13347 TT +CT genotypes are associated with increasing expression of CD44 in AML, which has been identified as a poor prognostic factor for this disease (Table 1).9 Although the molecular mechanism of association between CD44 rs13347 SNP and AML is unclear, increasing expression of CD44 can result in increased adhesion of HSCs to BM matrix that impairs normal differentiation and accumulation of immature blasts in BM niches. The results of a study by Loeffler-Ragg et al. can be mentioned in support of this hypothesis, which indicated that CD44 expression levels were higher in patients with myelodysplastic syndrome (MDS) progressing to AML relative to healthy controls.10 These findings suggest that the use of new therapeutic protocols for targeting CD44 can be useful in the improvement of AML patients. Since the induction of HSCs departure from BM to PB is the most common method to collect these stem cells for hematopoietic stem cell transplantation (HSCT), changing CD44 expression can be of high importance in stem cell mobilization. In this regard, investigations have shown that the TT genotype induced by CD44 rs13347 SNP is associated with increased expression of this marker, resulting in weak mobility of HSCs in response to granulocyte-colony stimulating factor (G-CSF) among patients with lymphoma and AML.11Martin-Antonio et al. indicated that healthy donors carrying the C allele of CD44 (CD44-2392 C) had lower expression of CD44 and increasing HSCs mobilization from BM to PB.12 Obviously, reaching a sufficient quantity of HSCs is an essential prerequisites for a successful transplantation; nevertheless, factors such as TT genotypes of CD44 rs13347 SNP that have been relatively ignored may be a factor of not achieving sufficient levels of HSCs in PB of patients or donors in both autologous or allogeneic transplants. Although quantitative evaluation of CD34+ HSCs by flow cytometry is the most frequent method to assess the sufficient number of HSCs, assessment of CD44 SNPs seems to be effective in predicting the efficacy of hemapheresis and reducing its episodes. Moreover, this approach may be helpful in decreasing the dose of G-CSF and its complications in both patients and healthy donors.

Table 1.

Genetic polymorphisms involved in clinicopathological characteristics of leukemia/lymphoma.

CD markers Chr. Biologic role SNP Region/Allele Disease Population Method Effect Ref.
CD33 19q13.4 Sialic acid-binding receptor rs12459419 Exon 2 C>T; Ala14Val AML American PCR Higher OS and decreased expression of CD33 4
rs1803254 3'-UTR G>C
rs2455069 (A>G); Arg69Gly AML     Higher relapse risk and low OS  
CD44 11p13 Involved in cell-cell interactions, cell adhesion and migration rs35112940 Exon 6 (AA+ AG)
CD185 11q23.3 Involved in B-cell migration into B-cell follicles of spleen and Peyer’s patches rs13347 promoter AML Chinese Western Blot May be a genetic modifier for developing AML 9
HL, NHL, AML Poland PCR Associated with poor mobilization of HSCs 11
rs6421571 Intron NHL Chinese PCR–RFLP Increased susceptibility to NHL and development of disease toward high Ann Arbor stage (III + IV) stage 14
rs80202369 Associated with increased susceptibility to NHL
rs1790192 Intron NHL, FL, DLBCL, Iowa PCR Associated with increased risk of malignancies and better event-free survival of FL patients. 15
PTCL

AML, acute myeloid leukemia; OS, overall survival; FL, follicular lymphoma; DLBCL, diffuse large B-cell lymphoma; PTCL, peripheral T-cell lymphoma; HL, Hodgkin lymphoma; NHL, non- Hodgkin lymphoma; HSC, hematopoietic stem cells; PCR, polymerase chain reaction; PCR-RFLP, polymerase chain reaction-restriction fragment length polymorphism.

CD185

CXCR5 or CD185 is a surface marker of circulating B-cells and follicular T-helper cells and plays an essential role in directing these cells toward secondary lymph nodes. The expression of CD185 has been reported in a number of malignancies, including non-Hodgkin lymphoma (NHL), which may indicate the involvement of this surface receptor in the development of these malignancies. 13 In addition, genetic studies have indicated the association between several polymorphisms of CD185 gene with NHL incidence (Table 1).14,15 The results of these investigations show that the percentage of CD185 rs6421571 CT, TT, and rs80202369 AA genotypes is higher in NHL patients than in healthy subjects. Those who carry the rs6421571 TT genotype show increasing tumor mass (≥5 cm vs. ˃5 cm) and progression to higher clinical stages of the disease.14 Interestingly, genetic research on the Iowa population has indicated that rs1790192 SNP of CD185 is related to an increased risk of follicular lymphoma (FL), while such patients have better survival than those without this SNP.15 This genetic change may account for transient and spontaneous improvement of FL patients that is seen in 25% of patients. Nevertheless, the hypothesis proposed by researchers for the association between CD185 SNPs and the incidence of NHL involves the impairment of activation and improper infiltration of lymphocyte lineages in lymph nodes as well as inhibition of tumor cell apoptosis.14 Given that extranodular tissues (including abdominal tissue) are of particular importance in lymphomas, CD185 SNPs may be an underlying cause of increasing extension and infiltration of tumor masses into these areas. Furthermore, disrupted function and migration of lymphocytes could reduce the resistance of patients to viral and bacterial infections, including Epstein-Barr virus (EBV), cytomegalovirus (CMV), and Helicobacter pylori (H. pylori), which are the most common factors of infection in patients with lymphoma.16,17

CD95/CD178

CD95 (Fas) induces apoptosis in cells following its binding to CD178 (Fas ligand) and the association between the presence of SNPs in the gene of this CD marker and clinical findings of AML patients has been demonstrated.18 CD95-1377 G>A polymorphism, often expressed as heterozygote (GA) and homozygote (AA) variants, is a known SNP among AML patients. This SNP has been shown to affect the binding site of CD95 with stimulatory protein 1 (SP1) transcription factor. Indeed, CD95-1377 G>A SNP generates a variant of CD95 gene that reduces the expression of this surface marker due to low binding affinity to SP1.18 Since CD95/CD178 interaction has a potential role in apoptosis, the induction of resistance to apoptosis of AML blast cells by these variants is a hypothesis supported by most researchers for the link between CD95-1377 G>A SNP and AML prognosis. -670 CD95 SNP is another polymorphism affecting CD95 binding to signal transducer and activator of transcription 1 (STAT1). Remarkably, allelic analysis of these SNPs showed that co-expression of - 1377A/-670A haplotype increased the risk of AML up to six-fold relative to those having -1377G/1377G haplotype.18 However; evidence suggests that the effects of CD95/CD178 SNPs are not similar in all populations. For example, no correlation has been reported between CD95/CD178 SNPs and AML incidence in Korean and Chinese populations.19,20 In fact, it is inferred that CD95/CD178 may have different functions in various populations depending on the inherited allele. Furthermore, SNPs could modulate protective or resistance mechanisms of AML blast cells against apoptosis by altering the function of downstream genes of CD95/CD178 promoter. Although no association has been reported between CD95-1377 SNP and chronic myeloid leukemia (CML), research has shown that -670 GG and CD178 −844 TC SNPs are associated with increasing risk of CML and disease progression to accelerated and blast phases.21 Unlike what observed in AML and CML, Tong et al. demonstrated that children carrying CD95 (-1377A, -670G) and CD178 (-844C) polymorphisms had reduced risk of acute lymphoblastic leukemia (ALL).22 Nonetheless, investigation of the association between these SNPs with ALL prognosis has shown that CD95-670 GG genotypes are directly related to liver tissue involvement in patients.23 Notably, Faree et al. in their study showed that CD95-670 AA genotype was associated with increased risk of incidence, more clinical findings, and decreased survival in patients with adult T-cell leukemia (ATL).24 In effect, these findings suggest that the association between CD95 SNPs is not limited to a specific genotype and that the effect of CD95-670 SNP on clinical findings of hematological malignancies appears to be greater than CD95-1377 SNP.

A notable point in the association between CD95/CD178 SNPs for prognosis of hematological malignancies is the difference in the effect of these genetic changes among diverse populations (Table 2).25,26 These contradictory results suggest that different effects of CD95/CD178 SNPs on the incidence and prognosis of hematological malignancies in various populations can be due to the influence of several factors, including environmental factors, race of population under survey, environmental-genetic interactions, frequency of alleles at these gene locations and the relationship between different genotypes derived from them. It seems that the biological roles of CD95/CD178 SNPs and their fixation in the genetic pool of individuals are of prognostic value. Hence, a general overview of the status of these polymorphisms and determining their relationship with the incidence and prognosis of hematological malignancies require extensive investigations in different populations of the world as well as comparison of results.

CD38

CD38 is a diagnostic marker of chronic lymphoblastic leukemia (CLL) that plays a vital role in activating intracellular signaling pathways following the stimulation of malignant B-cells receptors. CD38 expression is typically associated with increasing proliferation and survival of malignant B-cells and has been recognized as a poor prognostic marker in CLL.27,28 In addition, evidence has shown that CD38 is a prognostic marker that it increased expression is associated with B-CLL pathogenesis.29 rs6449182 SNP is a well-known polymorphism in CD38 gene and several studies have suggested its relationship with increased expression of CD38 and the risk of CLL in different populations (Table 3).30,31 Investigations on this polymorphism indicate that patients carrying the rs6449182 GG genotype have a higher expression level of CD38 and progression to higher clinical stages relative to patients lacking this genetic change.30-32 Although determining disease stage is the most common method for detecting CLL prognosis, clinical factors and laboratory findings such as age, sex, tumor mass, serum lactate dehydrogenase (LDH), and genetic background of patients are also of importance in assessment of disease progress. In this regard, Aydin et al. in a study on an Italian population showed that male CLL patients bearing the CD38 rs6449182 G allele had more aggressive disease than female patients.32 Although the overall risk of CLL is higher in older men than in women, the reason why men with CD38 rs6449182 G allele exhibit more aggressive disease has not been determined; this event may be related to decreasing production of sex hormones in older ages or the influence of environmental factors such as smoking that is an unfavorable environmental risk factor in men. Moreover, researchers found that the serum level of LDH as an indicator of tumor mass and disease progression to Richter syndrome (RS) was higher in patients with this genotype.32 Although the survival of CLL patients not having poor prognostic factors is approximately equal to normal individuals of similar age range, multiple genetic and environmental factors such as aging and the incidence of SNPs can alter disease progression conditions.33 For instance, elderly patients with CLL have been shown to bear increased risk of high cholesterol levels and atherosclerosis than younger patients.34 Interestingly, the results of Abramenko et al. research on a Ukrainian population have indicated that CD38 rs6449182 SNP is associated with increasing levels of total cholesterol (TC), triglycerides (TG), and low-density lipoproteins (LDL) in patients with CLL.31 Since the increase in LDL levels has been recognized as a trigger for neoplastic transformation of B-cells,35 CD38 rs6449182 SNP seems to be a poor prognostic factor increasing the risk of CLL in patients. In support of this hypothesis, there is evidence showing that LDL and HDL levels are elevated in the early stages of newly diagnosed CLL patients.34

Although CD38 expression is an essential factor in the prediction of CLL prognosis, evaluation of the impact of genetic changes on the expression of this marker and its relationship with clinical findings and response to treatment in patients has been relatively neglected. In brief, the detection of CD38 SNPs in the early phase of disease can provide valuable information on CLL prognosis and even the choice of appropriate treatment protocols independent of cytogenetic changes and clinical findings such as adenopathy and splenomegaly.

Table 2.

Summary of CD95/CD178 polymorphisms in hematological malignancies.

Type of malignancy Evaluated SNPs Number of Case/ Control Method Results Ref.
CD95 CD178
rs2234767 rs1800682 rs763110
(-1377G>A) (-670T>C) (-844T>C)
AML United Kingdom - 454/934 PCR-RFLP - CD95-1377AA has twice AML development risk. - 1377A/-670A haplotype of CD95 promoter SNP had highest risk of AML development. 18
Korea 592/858 PCR - Not associated with AML risk in the Korean population 19
China - - 98/2014 Meta-Analysis - Was not associated with AML developing. 20
Childhood de novo AML Multi-Ethnicity - - 440/ Not mentioned PCR - Reduction in CD95 enriches the expression of apoptosis resistant clones in AML blast cells. 25
Childhood ALL China - 361/519 PCR-RFLP - Combination of some protection variant forms is associated with decreased risk of childhood ALL 22
ALL Iran - 142/117 PCR-RFLP - –670 GG genotype is associated with liver involvement. 23
CML India - - 386/386 PCR-RFLP - CD95 −670 GG and CD178 −844 TC SNPs are associated with reduced event-free survival and development of CML
- CD95-670 AG associated with accelerated phase
- CD178-844TC SNP associated with development of blast phase
21
ATL Brazil - - 31/33 PCR-RFLP - Associated with susceptibility, clinical manifestation and survival. 24
FL Spain - - Cohort, 126 PCR - The C allele in this site is associated with a better response to rituximab therapy 26

AML, Acute myeloid leukemia; ALL, Acute lymphoblastic leukemia; CML, Chronic myeloid leukemia; ATL, adult T leukemia; FL, follicular lymphoma; SNP, single nucleotide polymorphism; PCR, polymerase chain reaction; PCR-RFLP, polymerase chain reaction-restriction fragment length polymorphism.

Table 3.

Association of CD38 polymorphisms and prognosis of CLL.

First author, year of study Country Case/Control Method Evaluated SNPs Results Refs
Jamroziak, 2009 Polish Caucasian 460/503 RFLP-PCR, ARMS-PCR, Flow cytometry rs6449182 rs1800561 - rs6449182 G and rs1800561 T alleles are associated with high expression of CD38 as well as increased risk of B-CLL. 30
Aydin, 2008 Italy 248/232 PCR, FACS, FISH rs6449182 - In combination analysis, the patient with rs6449182 G allele harboring high level of ZAP70 expression has unfavorable prognostic marker.
- rs6449182 G allele was associated with clinical and laboratory markers including increased LDH level, higher lymph node involvement and splenomegaly.
- rs6449182 G allele may represent an independent risk factor for RS development.
32
Abramenko, 2012 Ukrainian 328/271 RFLP-PCR rs6449182 - GG genotype is associated with dyslipidemia such as increase in LDL which is an antigenic stimulus for B-cells prior to or after neoplastic transformation. 31

PCR, polymerase chain reaction; PCR-RFLP, polymerase chain reaction-restriction fragment length polymorphism; FACS, fluorescence-activated cell sorter; FISH, fluorescence in situ hybridization; LDL, low density lipoprotein; LDH, lactate dehydrogenase; CLL, chronic lymphocytic leukemia; RS, Richter syndrome.

CD markers SNPs and defective of immune responses

Neutropenia is one of the most important side effects of persistent chemotherapy in hematological malignancies, which predisposes to bacterial and fungal infections. CD284 or toll-like receptor 4 (TLR4) is a surface marker of neutrophils interacting with lipopolysaccharides and activating the nuclear factor kB (NFkβ) signaling pathway, which plays a crucial role in combating pathogens and inhibiting apoptosis of neutrophils. Nonetheless, genetic investigations have shown that SNPs can cause neutropenia and recurrent infections in patients with hematological malignancies by impairing CD284 function. For example, three SNPs including rs11536889, rs1927911, and rs6478317 in CD284 gene, have been reported as a contributing factor to neutropenia and increased mortality in children with ALL.36 Considering the fact that drug intoxication is a complication of repeated chemotherapy in AML, the expression of CD284 SNPs can be associated with more serious consequences in these patients. In this regard, studies show that patients with AML who carry CD284 rs4986791 and rs4986790 SNPs as well as CD282 (TLR2) rs5743708 SNP are faced with exacerbated side effects of chemotherapy such as prolonged periods of fever and increased risk of pneumonia, invasive fungal disease, and sepsis.37,38 On the other hand, since HSCT is the most effective approach to achieve long-term remission in patients with AML, the incidence of CD284 SNPs might be a barrier to successful transplantation and a cause of relapse. Therefore, it seems that monitoring of these genetic changes in AML patients offers new approaches to the choice of supportive treatments after chemotherapy and HSCT in these patients. Studies indicate that decreased expression of CD284 is associated with lower resistance to lipopolysaccharides of pathogens, which increases susceptibility to H. pylori infection in individuals. Mucosa-associated lymphoid tissue (MALT) lymphoma is a type of lymphoma that originates from gastric tissue and is associated with H. pylori infection. Moreover, genetic studies on a German population have revealed that individuals carrying at least one G allele caused by rs4986790 SNP CD284 are more susceptible to H. pylori infection and progression of MALT lymphoma than others.39,40 Although no such association has been reported in the Greek population, evidence suggests a correlation between CD14-159C/T SNP and the incidence of MALT lymphoma in this population.41 Because CD284 acts as a transmembrane co-receptor for CD14 in response to microbial lipopolysaccharides, this type of SNP may be associated with impaired CD284/CD14 interaction. On the other hand, overexpression of CD14 has been recognized as a factor of body’s susceptibility to H. pylori and the initiation of inflammatory processes. 42 Given that autoimmune and inflammatory disorders such as Sjögren syndrome, Hashimoto’s thyroiditis, and ocular inflammation are common in some patients with MALT lymphoma,43 CD284 and CD14 SNPs might be involved in progression to these disorders. Besides, the importance of CD14 SNPs in the development and progression of MALT lymphoma could be related to the effect of these genetic changes on the function of B-cells as the main cells involved in this malignancy. As reported by Yu et al., CD14-260 TT genotypes are strongly associated with B-ALL expression, although such association has not been observed in TALL. 44

Inappropriate activation of TLRs has been reported as another underlying mechanism for the incidence of lymphoma. For instance, overexpression of CD289 (TLR9) is one of the most well-known abnormal expression patterns of TLRs in a wide range of lymphomas, including mantle cell, B-cell small lymphocytic, follicular, and diffuse large B-cell. In addition, the role of CD289 in binding unmethylated CpG DNA motifs of B-cells has been recognized to activate these cells against fungal, viral, and bacterial infections. CD289 can also be involved in the recognition of self nucleic acids and thus it is believed that increasing expression of CD289 is involved in the pathogenesis of autoimmune diseases.45 rs5743836 SNP is one of the most prevalent genetic alterations of CD289 that has been shown to increase susceptibility to HL and Crohn’s disease.46-48 Since rs5743836 SNP is associated with increased expression of CD289, genetic investigations have reported higher B-cell proliferation and subsequent enhancement of immune stimulation as a basic mechanism of this polymorphism in HL and NHL pathogenesis.47,49 However, the effect of this genetic variation is different across populations, so that rs5743836 SNP is related with increased NHL risk in the population of Portugal and Italy but no such association has been reported in the United States.49 Noack et al. recently showed that rs5743836 SNP can be effective in response to CD289 agonists, which is a therapeutic strategy inducing the death of Burkitt’s lymphoma (BL) cells (Table 4). Indeed, this finding suggests that CD289 rd5743836 SNP can be considered as a prognostic factor of response to treatment. 50 The importance of CD289 SNPs is not limited to the incidence and prognosis of lymphomas and the inheritance of different variants of CD289 can also affect different aspects of leukemia. Research has shown that patients with AML bearing CC genotype of rs5743836 have a higher overall survival after HSCT than those having TC/TT genotype.51 The CC allele of rs5743836 SNP appears to decrease CD289 expression, which is in turn associated with reduced activation of T helper and cytotoxic T-cells as well as decreasing graft versus host disease (GVHD),51 while rs352140 C variant allele is related with increasing incidence of viral infections in children with ALL who are in the remission phase after chemotherapy.52 It seems that polymorphisms in TLRs, as the first factors in detecting pathogens, play an essential role in determining susceptibility to infections and the occurrence of hematological malignancies. However, altered function of immune cells (including T-cells) is not dependent only on the expression and function of TLRs. Effective activation of T-cells requires a balance between stimulatory receptors such as CD28 and inducible co-stimulator (ICOS) or CD278 with inhibitory receptors like cytotoxic T-lymphocyte associated protein-4 (CTLA-4) or CD152. Impaired expression of these surface markers is related with the incidence of B-CLL.53,54 Besides, genetic research shows that CD152g.319C˃T, CD28c.17+3T˃C, and CD278c.1554+4GT SNPs play a role in the incidence of B-CLL in the Polish population. In addition, carriers of CD152g.319C,˃T,[T] and CD28c. 17+3T˃C SNPs are more likely to progress to higher stages of the disease.55 Progression of disease toward higher clinical stages in patients with CD278 (rs10932029) ISV1 + 173T>C [TT], (rs10183087) 602A>C [AA], (rs10932037) 1624C>T[CC], and (rs4675379 2373G>C [GG] has been reported to be lower than in other individuals.56 It is concluded that these genetic alterations change the expression status of T-cells by affecting the expressions of these CD markers (i.e. increasing expression of CD28, CD272, and CD278) in a way that sends stimulatory messages to B-cells and increases their proliferation. The precise mechanism of SNPs role in stimulatory and inhibitory T-cell receptors for the development and progression of hematological malignancies remains to be elucidated and needs further studies.

Effect of CD marker polymorphisms on targeted therapy

Although cytogenetic changes at the time of leukemia diagnosis (especially AML) are important prognostic factors, abnormal expression of genes due to mutation or SNPs can affect clinical findings and response to therapy. Perhaps homozygous inheritance of TT genotype of CD33 rs12459419 SNP is a molecular abnormality involved in response of AML patients to GO treatment. The carriers of this allele have a poor response to GO and show increasing minimal residual disease (MRD) levels.57,58 whereas those harboring the CC genotype of this polymorphism have higher diseasefree survival and reduced relapse risk following GO therapy;59 however, this prognostic pattern is not the same in all patients. Mortland et al. revealed that the TT genotype of rs12459419 was associated with a favorable-risk disease and good response to GO therapy, while patients carrying variant allele (AA+ AG) for CD33 rs35112940 have reduced overall and disease-free survival as well as increased risk of relapse.60 The remarkable variation in the effects of CD33 SNPs on clinical findings and response to treatment in AML patients may suggest that SNPs are an underlying factor of resistance to GO therapy in AML patients. Therefore, genetic sequencing of patients to detect the genetic changes is important from a biological point of view and can provide independent prognostic information on the identification of risk factors. Interestingly, the study of the effect of rs12459419 SNP on CD33 expression has yielded inconsistent results and has been shown to be associated with the decrease, increase, or lack of change in CD33 expression.58,59,61 These findings suggest that although CD33 is a unique prognostic marker in AML, SNPs might be more valuable than CD33 expression in the prognosis of disease progression and multidrug resistance (MDR). In other words, the diagnosis of this genetic disorder can be a differentiating factor in the prognosis of response to treatment among patients.

Table 4.

Polymorphisms of immune-related CD markers and prognostic effects in hematological malignancies.

CD markers Chr. Annotation name Function SNP Location Malignancy Method Effect Ref.
CD284 9q33.1 TLR4 Plays a fundamental role in pathogen recognition and activation of innate immunity rs11536889 rs1927911 3 ׳-UTR Intron ALL PCR Association with the risk of developing neutropenia and infectious complications 36
rs6478317 rs4986790 Promoter Exon 5 AML PCR Associated with long-lasting fever and increased risk of sepsis and pneumonia after chemotherapy 37
rs4986791 rs4986790 Promoter Exon 5 MALT lymphoma, HL Taq-Man Is one factor in the genetic susceptibility to gastric lymphoma 39,40
CD282 4q31.3 TLR2 Plays a fundamental role in pathogen recognition and activation of innate immunity rs5743708 Promoter AML PCR Associated with development of Sepsis, pneumonia, and invasive fungal disease in AML patients who undergo induction chemotherapy. 37,38
CD14 5q31.3 CD14 molecule Mediates the innate immune response to bacterial lipopolysaccharides –159C/T Promoter MALT lymphoma RFLP-PCR Enhances CD14 expression that might contribute to the development of H. pylori associated malignancy 41
rs2569190 Promoter ALL PCR Associated with developing risk of B-ALL 44
CD289 3p21.2 TLR9 Plays a fundamental role in pathogen recognition and activation of innate immunity. rs5743836 Promoter HL PCR May increase the susceptibility to HL 48
NHL PCR Enhanced CD289 function that increases B-cell proliferation upon CpG stimulation as well as NLH risk 49
BL qRT-PCR C allele results in enhanced NF-kB activation following TLR9 triggering that leads to induced apoptosis in BL cells 50
CD28 2q33.2 CD28 molecule Is essential for T-cell proliferation, activation, and survival rs352140 Promoter AML PCR C allele is associated with reduced CD289 gene expression and slower reconstitution of Th and cytotoxic T cells after transplantation 51
17+3T> Promoter ALL PCR C variant allele is associated with increased risk of infection during the remission induction period of treatment 52
CD278 2q33.2 ICOS Plays an important role in regulation of immune responses 1554+4GT Promoter B-CLL PCR CD28 and CD278 may be associated with dysregulation of immune response and increased proliferation of B- cells that leads to increased risk of B-CLL 55
CD152 2q33.2 CTLA-4 Transmits an inhibitory signal to T cells g319C>T +49A/G Promoter NHL PCR May have a role in genetic susceptibility to NHL 57

ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; B-CLL, B-chronic lymphoblastic leukemia; MALT, mucosa-associated lymphoid tissue; HL, Hodgkin lymphoma; NHL, Non Hodgkin lymphoma; BL, Burkitt's lymphoma; H. pylori, Helicobacter pylori; NF-kB, nuclear factor kB; TLR, toll like receptor; Th, T helper; ICOS, inducible co-stimulator; CTLA-4, cytotoxic T-lymphocyte associated protein-4; PCR, polymerase chain reaction; PCR-RFLP, polymerase chain reaction-restriction fragment length polymorphism.

CD20 is the most important marker on 80% of malignant Bcell masses in disorders such as CLL and diffuse large B-cell lymphoma (DLBCL). The expression of this marker increases over the course of malignancy and is a potential therapeutic target for rituximab (RTX), a chimerical monoclonal antibody against CD20 on B-cells and R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone). Rs2070770 is a common polymorphism in CD20 gene that has been reported to be associated with CLL.62 In addition, research has shown that SNPs can affect CD20 expression and response to RTX-containing drugs. For instance, Zang et al. revealed that patients with de novo DLBCL in a Chinese population carrying the T allele of CD20 rs2070770 SNP are associated with decreased CD20 expression, a favorable response to RTX-containing drugs, and increase in survival.63 However, the results of another study in the same population indicated that the subjects who inherit C genotype of this SNP have a better response to treatment and a longer survival than those with at least one T allele.64 While the precise mechanism of the influence of different alleles on response to RTX therapy has not been elucidated, the CD20 rs2070770 SNP could be a favorable prognostic factor in patients with DLBCL. However, there is evidence suggesting that it has no effect on DLBCL prognosis.65 These findings indicated that the association between CD20 SNP and RTXcontaining- drugs is a controversial topic and that determining the impact of these polymorphisms on disease prognosis requires further collaboration among researchers to obtain accurate information in this regard. The response to R-CHOP in DLBCL patients may not be related solely to genetic alterations of CD20 as the primary target of this therapeutic protocol but rather the transfer of drug components across the membrane (ABCG2) or CD338 ATP binding cassette could have been disrupted. ATP binding cassette subfamily G member 2 (ABCG2) or CD338 is a membrane pump that has been shown to increase the flow of drugs such as doxorubicin, vincristine, and prednisone from cytoplasm to the extracellular matrix.66 Moreover, CD338 is a major cause of MDR in hematological malignancies,67 so that targeting CD338 is widely used in DLBCL to overcome MDR.68 Liu et al. in a recent study revealed that patients with DLBCL treated with R-CHOP carrying CD338 rs2231137 GG genotype had poorer progression-free survival (PFS) and event-free survival (EFS) than those bearing AA or GG genotypes.69 It seems that the GG genotype could have produced a phenotype of CD338 that is unable to transport drug components across the membrane. There is also evidence suggesting that AA or GG genotypes are favorable factors reducing the risk of infection in patients with DLBCL treated with R-CHOP.70 Indeed, these findings indicate that the genetic background of patients can also be effective on clinical presentations of a particular therapeutic protocol.

Discussion and future perspective

Hematological malignancies are neoplastic disorders, and cytogenetic abnormalities as well as clinical features are the most important factors in their classification. The phenotype of the involved cells, often characterized by the expression of surface CD markers, is another feature playing an essential role in the diagnosis and prognosis of hematological malignancies and solid tumors.3,71,72 Although flow cytometry is a practical technique to analyze the expression levels of CD markers, genetic alterations (including SNPs) affecting expression patterns of CD markers may cause significant limitations in the classification and precise prognosis of hematological malignancies.73-75 As an example, CD38 rs6449182 is a known polymorphism in CLL and it has been shown that patients carrying the GG genotype of this polymorphism have more aggressive disease.30,31 Although clinical symptoms such as BM, PB, and lymph node involvement are the most crucial prognostic factors in CLL, the CD38 rs6449182 SNP has been shown to be associated with biochemical abnormalities such as increased LDH and lipoproteins, which indicates the progression toward advanced clinical stages. Since the increase in lipoproteins such as HDL and LDL has been identified as a stimulus for increasing B-cell proliferation.35 the evaluation of CD38 SNPs should provide new clues regarding the etiology and development of CLL. Similarly, research has shown that CD95-670 G>A and CD178 −844 T>C SNPs are associated with the progression of CML toward accelerated and blast phases.21 Given that CD95/CD178 is involved in the apoptosis process, these genetic changes could influence blast cells resistance to apoptosis by affecting the expression and alteration of intracellular mechanisms. Indeed, this finding suggests that CD95/CD178 SNPs as potential prognostic factors provide for different degrees of disease progression in CML patients and are important in defining high-risk groups.

Although the expression pattern of CD markers is not widely used in differential diagnosis of lymphomas, CD markers’ SNPs appear to be important in the prognosis of these disorders that are associated with extracellular infiltration of malignant cells. Conceivably, CD185 SNPs that are related with the infiltration of malignant lymphocytes into lymph nodes, increased tumor mass size, and progression to higher clinical stages in the NHL have been identified as a poor prognostic factor in this disorder.14 In fact; evaluation of SNPs in lymphomas can help determine the prognosis and the extent of tissue involvement. SNPs in CD markers, especially immune-related CD markers, have clinical implications and might lead to alleles of these functional surface markers that are not susceptible to infectious pathogens, paving the way for progression to malignancies. Similarly, several investigations have revealed that genetic variation from SNPs in immune-related CD markers such as CD284 and CD14 can provide conditions for H. pylori infection and the incidence of MALT lymphoma.39,41 It seems that CD284 and CD14 SNPs contribute to H. pylori survival in the acidic environment of the stomach and its confrontation with host’s immune system and thus have serious clinical consequences. However, epidemiological studies have shown that the effect of these SNPs is not the same in all populations, so no association was reported between these polymorphisms and the incidence of MALT lymphoma in the Greek population.41 In fact, it seems that various factors such as ethnicity and geography are involved in the extent of adaptation to genetic variation resulting from polymorphisms as well as susceptibility to H.pylori and progression toward malignancies. Because immune-related CD markers play an essential role in regulating immune function, imbalances in their expressions have been identified as a factor of body’s susceptibility to a variety of diseases. CD152g.319C˃T, CD28c.17+ 3T˃C, and CD278c.1554+4GT SNPs that cause imbalanced expressions of inhibitory and stimulatory T-cell receptors have been recognized as poor prognostic factors in association with increased risk of BCLL. 55,56 It seems that these SNPs decrease the activity of inhibitory receptor promoters (CD152) and in contrast increase the activity of the stimulatory receptors (CD28 and CD278), which results in increased proliferation of B-cells and susceptibility to B-CLL.55 However, the precise mechanism of these SNPs is unclear and needs further study.

One of the biggest challenges facing patients with hematological malignancies is the failure to respond favorably to treatment as well as disease relapse. The fact that many drugs have diverse effects on different individuals has been accepted, but there is evidence suggesting that patients often have different levels of resistance to drugs, some of them needing lower or higher doses to achieve the desired therapeutic effect and sometimes a medication may not have any therapeutic effect in patients. Although drug interactions can sometimes lead to poor response to therapy in patients using multiple drugs, many differences can be due to genetic variation. For instance, clinical results from genetic studies on AML and DLBCL patients have shown that different genotypes of CD20 rs2070770 SNP, as well as CD33 rs12459419 and rs35112940 SNPs, can be effective in the mode of patients’ response to treatment.58-60,63,64 Researchers also believe that the expressions of SNPs in proteins involved in the transport of drugs, which are often expressed as CD markers at the cell surface, can influence the response of the patients to many drugs.69 It is inferred that the genetic makeup of patients can be a main cause of variation in response to drugs. These findings point to the necessity of individual medical treatment, which simply means prescribing specific medications and treatments based on genetic makeup that works best for the patient and minimize failure of optimal response and risk of recurrence. Overall, the results of genetic investigations indicate that SNPs of CD markers are a main factor for altering and even determining the biological phenotype of hematological malignancies. However, SNPs by themselves cannot affect different aspects of these disorders, including incidence, progression, prognosis, and response to therapy by altering the expression and function of CD markers, but the interaction of these genetic changes with environmental factors determines the risk of disease in an individual, severity of the disease, and even how they respond to treatment. These findings suggest that the prognosis of SNPs in CD markers varies across populations and may even lead to different responses to therapeutic agents. In other words, some of these genetic changes can cause rapid progression of the disease and require a different type and dose of a particular drug in patients, while the same change could not have significant effect on individual or other populations. Although it is difficult to design genetic tests that can identify a particular SNP and predict its impact on a persons disease progression and response to a specific drug, it appears that the detection of genetic changes (including SNPs) in CD markers as significant diagnostic and prognostic factors in hematological malignancies can contribute to better understanding and predict the clinical course of these disorders. Moreover, physicians will be able to quickly prescribe optimal treatment with available drugs by analyzing the genetic history, maximizing the value of the treatment process by choosing a precise treatment protocol. It is hoped that the evaluation of SNPs of CD markers will enable early detection of these disorders, their prognosis and response to treatment besides assessing the potential for hematological malignancies in different populations around the world, thereby minimize side effects as well as the cost of medical care for patients by providing appropriate doses of drug.

Highlights

  • Several CD marker-related SNPs can affect leukemia/lymphoma pathogenesis and prognosis.

  • SNPs in multiple CD markers can be associated with drug resistance in leukemia/lymphoma.

  • Different CD marker-related SNPs can be a new therapeutic target in leukemia/lymphoma.

Acknowledgements

We wish to thank all our colleagues in Allied Health Sciences School, Ahvaz Jundishapur University of Medical Sciences.

References

  • 1.Zola H, Swart B, Nicholson I, et al. CD molecules 2005: human cell differentiation molecules. Blood 2005;106:3123-6. [DOI] [PubMed] [Google Scholar]
  • 2.Sarma A, Hazarika M, Das D, et al. Expression of aberrant CD markers in acute leukemia: a study of 100 cases with immunophenotyping by multiparameter flowcytometry. Cancer Biomark 2015;15:501-5. [DOI] [PubMed] [Google Scholar]
  • 3.Karai B, Habok M, Remenyi G, et al. A novel flow cytometric method for enhancing acute promyelocytic leukemia screening by multidimensional dot-plots. Ann Hematol 2019;98:1413-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Mortland L, Alonzo TA, Walter RB, et al. Clinical significance of CD33 nonsynonymous single-nucleotide polymorphisms in pediatric patients with acute myeloid leukemia treated with gemtuzumab-ozogamicin-containing chemotherapy. Clin Cancer Res 2013;19:1620-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Walter RB, Appelbaum FR, Estey EH, et al. Acute myeloid leukemia stem cells and CD33-targeted immunotherapy. Blood 2012;119:6198-208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mingari MC, Vitale C, Romagnani C, et al. p75/AIRM1 and CD33, two sialoadhesin receptors that regulate the proliferation or the survival of normal and leukemic myeloid cells. Immunol Rev 2001;181:260-8. [DOI] [PubMed] [Google Scholar]
  • 7.Fostvedt LK, Hibma JE, Masters JC, et al. Pharmacokinetic/Pharmacodynamic Modeling to Support the Re-approval of Gemtuzumab Ozogamicin. Clin Pharmacol Ther 2019;106:1006-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Williams K, Motiani K, Giridhar PV, et al. CD44 integrates signaling in normal stem cell, cancer stem cell and (pre) metastatic niches. Exp Biol Med 2013;238:324-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wu H, Deng J, Zheng J, et al. Functional polymorphisms in the CD44 gene and acute myeloid leukemia cancer risk in a Chinese population. Mol Carcinog 2015;54:102-10. [DOI] [PubMed] [Google Scholar]
  • 10.Loeffler-Ragg J, Germing U, Sperr W, et al. Serum CD44 levels predict survival in patients with low-risk myelodysplastic syndromes. Crit Rev Oncol Hematol 2011;78:150-61. [DOI] [PubMed] [Google Scholar]
  • 11.Szmigielska-Kaplon A, Szemraj J, Hamara K, et al. Polymorphism of CD44 influences the efficacy of CD34+ cells mobilization in patients with hematological malignancies. Biol Blood Marrow Transplant 2014;20:986-91. [DOI] [PubMed] [Google Scholar]
  • 12.Martin-Antonio B, Carmona M, Falantes J, et al. Impact of constitutional polymorphisms in VCAM1 and CD44 on CD34+ cell collection yield after administration of granulocyte colony-stimulating factor to healthy donors. Haematologica 2011;96:102-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Trentin L, Cabrelle A, Facco M, et al. Homeostatic chemokines drive migration of malignant B cells in patients with non- Hodgkin lymphomas. Blood 2004;104:502-8. [DOI] [PubMed] [Google Scholar]
  • 14.Song H, Tong D, Cha Z, et al. CXC chemokine receptor type 5 gene polymorphisms are associated with non-Hodgkin lymphoma. Mol Biol Rep 2012;39:8629-35. [DOI] [PubMed] [Google Scholar]
  • 15.Charbonneau B, Wang AH, Maurer MJ, et al. CXCR5 polymorphisms in non-Hodgkin lymphoma risk and prognosis. Cancer Immunol Immunother 2013;62:1475-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hussain SK, Zhu W, Chang SC, et al. Serum levels of the chemokine CXCL13, genetic variation in CXCL13 and its receptor CXCR5, and HIV-associated non-hodgkin B-cell lymphoma risk. Cancer Epidemiol Biomarkers Prev 2013;22:295-307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mehravaran H, Makvandi M, Samarbaf Zade A, et al. Association of Human Cytomegalovirus with Hodgkins Disease and Non-Hodgkins lymphomas. Asian Pac J Cancer Prev 2017;18:593-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sibley K, Rollinson S, Allan JM, et al. Functional FAS promoter polymorphisms are associated with increased risk of acute myeloid leukemia. Cancer Res 2003;63:4327-30. [PubMed] [Google Scholar]
  • 19.Kim HJ, Jin XM, Kim H-N, et al. Fas and FasL polymorphisms are not associated with acute myeloid leukemia risk in Koreans. DNA Cell Biol 2010;29:619-24. [DOI] [PubMed] [Google Scholar]
  • 20.Huang Y, Deng D, Li H, et al. Fas 670A> G polymorphism is not associated with an increased risk of acute myeloid leukemia development. Biomed Rep 2016;4:153-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Edathara PM, Gorre M, Kagita S, et al. Association of promoter polymorphisms of Fas-FasL genes with development of Chronic Myeloid Leukemia. Tumor Biol 2016;37:5475-84. [DOI] [PubMed] [Google Scholar]
  • 22.Tong N, Zhang L, Sheng X, et al. Functional polymorphisms in FAS, FASL and CASP 8 genes and risk of childhood acute lymphoblastic leukemia: a case–control study. Leuk Lymphoma 2012;53:1360-6. [DOI] [PubMed] [Google Scholar]
  • 23.Valibeigi B, Amirghofran Z, Golmoghaddam H, et al. Fas gene variants in childhood acute lymphoblastic leukemia and association with prognosis. Pathol Oncol Res 2014;20:367-74. [DOI] [PubMed] [Google Scholar]
  • 24.Farre L, Bittencourt A, Silva-Santos G, et al. Fas–670 promoter polymorphism is associated to susceptibility, clinical presentation, and survival in adult T cell leukemia. J Leukoc Biol 2008;83:220-2. [DOI] [PubMed] [Google Scholar]
  • 25.Mehta PA, Gerbing RB, Alonzo TA, et al. FAS promoter polymorphism: outcome of childhood acute myeloid leukemia. A children’s oncology group report. Chin Clin Oncol 2008;14:7896-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gutierrez-Civicos R, Hurtado AM, Torres-Moreno D, et al. A polymorphism in FASL is associated with rituximab response in follicular lymphoma patients. Am J Hematol 2016;91:E305-7. [DOI] [PubMed] [Google Scholar]
  • 27.Deaglio S, Vaisitti T, Serra S, et al. CD38 in chronic lymphocytic leukemia: from bench to bedside? Mini Rev Med Chem 2011;11:503-7. [DOI] [PubMed] [Google Scholar]
  • 28.Deaglio S, Vaisitti T, Zucchetto A, et al. CD38 as a molecular compass guiding topographical decisions of chronic lymphocytic leukemia cells. Semin Cancer Biol 2010;20:416-23. [DOI] [PubMed] [Google Scholar]
  • 29.Deaglio S, Vaisitti T, Aydin S, et al. In-tandem insight from basic science combined with clinical research: CD38 as both marker and key component of the pathogenetic network underlying chronic lymphocytic leukemia. Blood 2006;108:1135-44. [DOI] [PubMed] [Google Scholar]
  • 30.Jamroziak K, Szemraj Z, Grzybowska-Izydorczyk O, et al. CD38 gene polymorphisms contribute to genetic susceptibility to B-cell chronic lymphocytic leukemia: evidence from two case-control studies in Polish Caucasians. Cancer Epidemiol Biomarkers Prev 2009;18:945-53. [DOI] [PubMed] [Google Scholar]
  • 31.Abramenko IV, Bilous NI, Pleskach GV, et al. CD38 gene polymorphism and risk of chronic lymphocytic leukemia. Leuk Res 2012;36:1237-40. [DOI] [PubMed] [Google Scholar]
  • 32.Aydin S, Rossi D, Bergui L, et al. CD38 gene polymorphism and chronic lymphocytic leukemia: a role in transformation to Richter syndrome? Blood 2008;111:5646-53. [DOI] [PubMed] [Google Scholar]
  • 33.Keating MJ, Chiorazzi N, Messmer B, et al. Biology and treatment of chronic lymphocytic leukemia. ASH Educ Prog Book 2003;2003:153-75. [DOI] [PubMed] [Google Scholar]
  • 34.Yavasoglu I, Sargin G, Yilmaz F, et al. Cholesterol levels in patients with chronic lymphocytic leukemia. J Natl Med Assoc 2017;109:23-7. [DOI] [PubMed] [Google Scholar]
  • 35.Rosén A, Murray F, Evaldsson C, et al. Antigens in chronic lymphocytic leukemia-implications for cell origin and leukemogenesis. Sem Cancer Biol 2010;20:400-9. [DOI] [PubMed] [Google Scholar]
  • 36.Miedema K, Te Poele E, Tissing W, et al. Association of polymorphisms in the TLR4 gene with the risk of developing neutropenia in children with leukemia. Leukemia 2011;25:995. [DOI] [PubMed] [Google Scholar]
  • 37.Schnetzke U, Spies-Weisshart B, Yomade O, et al. Polymorphisms of Toll-like receptors (TLR2 and TLR4) are associated with the risk of infectious complications in acute myeloid leukemia. Genes Immun 2014;16:83. [DOI] [PubMed] [Google Scholar]
  • 38.Fischer M, Spies-Weisshart B, Schrenk K, et al. Polymorphisms of Dectin-1 and TLR2 predispose to invasive fungal disease in patients with acute myeloid leukemia. PLoS one 2016;11:e0150632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hellmig S, Fischbach W, Goebeler-Kolve M-E, et al. Association study of a functional Toll-like receptor 4 polymorphism with susceptibility to gastric mucosa-associated lymphoid tissue lymphoma. Leuk Lymphoma 2005;46:869-72. [DOI] [PubMed] [Google Scholar]
  • 40.Nieters A, Beckmann L, Deeg E, et al. Gene polymorphisms in Toll-like receptors, interleukin-10, and interleukin-10 receptor alpha and lymphoma risk. Genes Immun 2006;7:615-24. [DOI] [PubMed] [Google Scholar]
  • 41.Tuere-Oezdemir F, Gazouli M, Tzivras M, et al. Association of polymorphisms of NOD2, TLR4 and CD14 genes with susceptibility to gastric mucosa-associated lymphoid tissue lymphoma. Anticancer Res 2008;28:3697-700. [PubMed] [Google Scholar]
  • 42.Schmaußer B, Andrulis M, Endrich S, et al. Toll-like receptors TLR4, TLR5 and TLR9 on gastric carcinoma cells: an implication for interaction with Helicobacter pylori. Int J Med Microbiol 2005;295:179-85. [DOI] [PubMed] [Google Scholar]
  • 43.Zullo A, Hassan C, Ridola L, et al. Gastric MALT lymphoma: old and new insights. Ann Hepatol 2014;27:27-33. [PMC free article] [PubMed] [Google Scholar]
  • 44.Yu X, Zhang C, Sun A, et al. Genetic variations in CD14 promoter and acute lymphoblastic leukemia susceptibility in a Chinese population. DNA and Cell Biol 2011;30:777-82. [DOI] [PubMed] [Google Scholar]
  • 45.Marshak-Rothstein A. Toll-like receptors in systemic autoimmune disease. Nat Rev Immunol 2006;6:823-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hong J, Leung E, Fraser AG, et al. TLR2, TLR4 and TLR9 polymorphisms and Crohn’s disease in a New Zealand Caucasian cohort. J Gastrointest Hepatol 2007;22:1760-6. [DOI] [PubMed] [Google Scholar]
  • 47.Mollaki V, Georgiadis T, Tassidou A, et al. Polymorphisms and haplotypes in TLR9 and MYD88 are associated with the development of Hodgkins lymphoma: a candidate-gene association study. J Hum Genet 2009;54:655. [DOI] [PubMed] [Google Scholar]
  • 48.Rahman HAA, Khorshied MM, Khorshid OMR, et al. Toll-like receptor 2 and 9 genetic polymorphisms and the susceptibility to B cell non-Hodgkin lymphoma in Egypt. Anna Hematol 2014;93:1859-65. [DOI] [PubMed] [Google Scholar]
  • 49.Carvalho A, Cunha C, Almeida AJ, et al. The rs5743836 polymorphism in TLR9 confers a population-based increased risk of non-Hodgkin lymphoma. Genes Immun 2012;13:197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Noack J, Jordi M, Zauner L, et al. TLR9 agonists induced cell death in Burkitt’s lymphoma cells is variable and influenced by TLR9 polymorphism. Cell Death Amp Dise 2012;3:e323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Elmaagacli AH, Steckel N, Ditschkowski M, et al. Toll-like receptor 9, NOD2 and IL23R gene polymorphisms influenced outcome in AML patients transplanted from HLA-identical sibling donors. Bone Marrow Transplant 2010;46:702. [DOI] [PubMed] [Google Scholar]
  • 52.Bhadri VA, Beckett SM, Duncan C, et al. Variation in Toll-like receptor 9 gene modifies the risk of infection in children treated for acute lymphoblastic leukemia. Leuk Lymphoma 2012;53:1828-30. [DOI] [PubMed] [Google Scholar]
  • 53.Frydecka I, Kosmaczewska A, Bocko D, et al. Alterations of the expression of T-cell-related costimulatory CD28 and downregulatory CD152 (CTLA-4) molecules in patients with B-cell chronic lymphocytic leukaemia. Br J Cancer 2004;90:2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Motta M, Rassenti L, Shelvin B, et al. Increased expression of CD152 (CTLA-4) by normal T lymphocytes in untreated patients with B-cell chronic lymphocytic leukemia. Leukemia 2005;19:1788. [DOI] [PubMed] [Google Scholar]
  • 55.Suwalska K, Pawlak E, Karabon L, et al. Association studies of CTLA-4, CD28, and ICOS gene polymorphisms with B-cell chronic lymphocytic leukemia in the Polish population. Human Immunol 2008;69:193-201. [DOI] [PubMed] [Google Scholar]
  • 56.Karabon L, Jedynak A, Tomkiewicz A, et al. ICOS gene polymorphisms in B-cell chronic lymphocytic leukemia in the Polish population. Folia Histochem Cytobiol 2011;49:49-54. [DOI] [PubMed] [Google Scholar]
  • 57.Monne M, Piras G, Palmas A, et al. Cytotoxic T-lymphocyte antigen-4 (CTLA-4) gene polymorphism and susceptibility to non-Hodgkins lymphoma. Am J Hematol 2004;76:14-8. [DOI] [PubMed] [Google Scholar]
  • 58.Lamba JK, Pounds S, Cao X, et al. Coding polymorphisms in CD33 and response to gemtuzumab ozogamicin in pediatric patients with AML: a pilot study. Leukemia 2008;23:402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lamba JK, Chauhan L, Shin M, et al. CD33 splicing polymorphism determines gemtuzumab ozogamicin response in de novo acute myeloid leukemia: report from randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol 2017;35:2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mortland L, Alonzo TA, Walter RB, et al. Clinical Significance of CD33 Nonsynonymous Single-Nucleotide Polymorphisms in Pediatric Patients with Acute Myeloid Leukemia Treated with Gemtuzumab-Ozogamicin-Containing Chemotherapy. Clin Cancer Res 2013;19:1620-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Malik M, Chiles J, III, Xi HS, et al. Genetics of CD33 in Alzheimers disease and acute myeloid leukemia. Hum Mol Genet 2015;24:3557-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Fang C, Zhu D-X, Wang L, et al. Association between polymorphism of CD20 gene and chronic lymphocytic leukemia in Chinese population. Int Clin Eexper Med 2015;8:11235. [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhang L-N, Wang L, Fang C, et al. The significance of single nucleotide polymorphism rs2070770 in CD20 gene in Chinese patients with diffuse large B-cell lymphoma. Leuk lymphoma 2015;56:676-81. [DOI] [PubMed] [Google Scholar]
  • 64.Ding H, Jin X, Ding N, et al. Single nucleotide polymorphisms of CD20 gene and their relationship with clinical efficacy of RCHOP in patients with diffuse large B cell lymphoma. Cancer cell Int 2013;13:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sar A, Perizzolo M, Stewart D, et al. Mutation or polymorphism of the CD20 gene is not associated with the response to R-CHOP in diffuse large B cell lymphoma patients. Leuk Res 2009;33:792-7. [DOI] [PubMed] [Google Scholar]
  • 66.Cascorbi I. Role of pharmacogenetics of ATP-binding cassette transporters in the pharmacokinetics of drugs. Pharmacol Therap 2006;112:457-73. [DOI] [PubMed] [Google Scholar]
  • 67.Shahrabi S, Behzad MM, Jaseb K, et al. Thrombocytopenia in leukemia: Pathogenesis and prognosis. Histol Histopathol 2018;33:895-908. [DOI] [PubMed] [Google Scholar]
  • 68.Singh RR, Kunkalla K, Qu C, et al. ABCG2 is a direct transcriptional target of hedgehog signaling and involved in stroma- induced drug tolerance in diffuse large B-cell lymphoma. Oncogene 2011;30:4874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liu D, Wu N, Sun H, et al. ABCG2 and NCF4 polymorphisms are associated with clinical outcomes in diffuse large B-cell lymphoma patients treated with R-CHOP. Oncotarget 2017;8:58292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rossi D, Rasi S, Franceschetti S, et al. Analysis of the host pharmacogenetic background for prediction of outcome and toxicity in diffuse large B-cell lymphoma treated with RCHOP21. Leukemia 2009;23:1118. [DOI] [PubMed] [Google Scholar]
  • 71.Shahrabi S, Ehsanpour A, Heidary S, et al. Expression of CD markers in JAK2V617F positive myeloproliferative neoplasms: Prognostic significance. Oncol Rev 2018;12:373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Rezaeeyan H, Shahrabi S, McKee TD, et al. The expression of CD markers in solid tumors: Significance in metastasis and prognostic value. Histol Histopathol 2018;33:1005-12. [DOI] [PubMed] [Google Scholar]
  • 73.Asnafi AA, Behzad MM, Ghanavat M, et al. Singe nucleotide polymorphisms in osteosarcoma: Pathogenic effect and prognostic significance. Exper Molecul Pathol 2019;106:63-77. [DOI] [PubMed] [Google Scholar]
  • 74.Ghanavat M, Ebrahimi M, Rafieemehr H, et al. Thrombocytopenia in Solid Tumors: Prognostic Significance. Oncol Rev 2019;13:413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Asnafi AA, Farshchi N, Khosravi A, et al. Significance of genetic polymorphisms in hematological malignancies: implications of risk factors for prognosis and relapse. MEMO 2018;11:330-44. [Google Scholar]

Articles from Oncology Reviews are provided here courtesy of Frontiers Media SA

RESOURCES