Abstract
Cancer biomarkers have provided great opportunities for improving the management of cancer patients by enhancing the efficiency of early detection, diagnosis, and efficacy of treatment. Every cell type has a unique molecular signature, referred to as biomarkers, which are identifiable characteristics such as levels or activities of a myriad of genes, proteins, or other molecular features. Biomarkers can facilitate the molecular definition of cancer, provide information about the course of cancer, and predict response to chemotherapy. They offer the hope of early detection as well as tracking disease progression and recurrence. Current progress in the characterization of molecular genetics of HIV-associated cancers may form the basis for improved patient stratification and future targeted or individualized therapies. Biomarker use for cancer staging and personalization of therapy at the time of diagnosis could improve patient care. This review focuses on the relevance of biomarkers in the most common HIV-associated malignancies, namely, Kaposi sarcoma, non-Hodgkin’s lymphoma, and invasive cervical cancer.
Keywords: biomarkers, cancer, HIV, non-Hodgkin’s lymphoma, Kaposi sarcoma, cervical cancer
Introduction
Cancer is a genetically and clinically diverse disease, whose pathogenesis, aggressiveness, metastatic potential, and response to treatment can be different among individual patients.1 Great variations exist, even between individuals with the same type of cancer, suggesting the role of genetic factors in cancer pathogenesis. The risk of developing cancer is greatly increased in human immunodeficiency virus (HIV) setting, and it is increasingly recognized as a complication of HIV infection.2,3 Cancers with an increased incidence in HIV patients include the AIDS-defining malignancies [Kaposi’s sarcoma, non-Hodgkin’s lymphoma (NHL), and invasive cervical cancer] and other non-AIDS-defining cancers (Hodgkin’s lymphoma, hepatocellular carcinoma, and lung cancer).4 Due to the complexity and diversity of cancer, the application of personalized medicine in the management of cancer patients has been suggested and encouraged.
Personalized medicine hinges on biomarkers, which are highly sensitive and specific in revealing information that is relevant for diagnosis, prognosis, and therapy.5,6 Thus, biomarker discovery and development are one of the cores of personalized medicine for cancer. Cancer biomarkers may be discovered using molecular, cellular, and imaging methodologies focused on drug and disease mechanisms, thus providing critical feedback about the interaction of novel therapies with their intended target and about the disease itself.7 Biomarkers play a role in cancer screening, early diagnosis, prognosis, cancer stratification, prediction of treatment efficacy, and adverse reaction. A biomarker can consist of genomic and proteomic patterns, single genes or proteins, chromosomal abnormalities, epigenetic signatures, aberrant microRNA (miRNA), as well as imaging changes observed on magnetic resonance imaging (MRI) or positron emission tomography (PET) scan. However, most biomarkers have both prognostic and predictive value.
Biomarker Definition
Biomarkers are characteristics that are objectively measured and evaluated as indicators of normal biological processes, pathogenic processes, and pharmacological responses to a therapeutic intervention.8,9 In cancer, biomarkers are defined as biochemical substances elaborated by cancer cells either due to the cause or effect of malignant process.10 However, cancer biomarkers must be detectable only in the presence of cancer. Cancer biomarkers may be detected in sample matrices such as serum, plasma, whole blood, urine, and tissue.11 They can be normal endogenous products that are produced at a greater rate in cancer cells or the products of newly switched on genes that remained inactive in normal cells. Biomarkers may include intracellular molecules or proteins in tissues or may be released into the circulation and appear in serum, and their presence in significant amount may indicate the presence of cancer. However, the usefulness of a biomarker lies in its ability to provide early indication of a disease or its progression, and it should be easy to detect and should be measurable across populations.12
Cancer Biomarker Classification and Utility
It has been well established that a variety of biomarkers are used in risk assessment, early detection, diagnosis, treatment, and management of cancer.13,14 Molecular analyses at the protein, DNA, RNA, or miRNA levels can contribute to the identification of novel tumor subclasses, each with a unique prognostic outcome or response to treatment.15 Biomarkers enable the characterization of patient populations and quantitation of the extent to which drugs reach intended targets, alter proposed pathophysiological mechanisms, and achieve clinical outcomes.16 The most valuable biomarkers are highly sensitive, specific, reproducible, and predictable, and the majority of US Food and Drug Administration (FDA) approved that cancer biomarkers are serum-derived single proteins.17,18
Biomarkers can be classified based on different parameters such as characteristics and function. Biomarkers that are classified according to their functions include type 0 biomarkers, which measure the natural history of a disease and they should correlate over time with known clinical indicators; type I biomarkers are associated with the effectiveness of pharmacologic agents; and type II biomarkers, also known as surrogate endpoint biomarkers, are intended to substitute for clinical endpoints.19 Current cancer biomarkers may be grouped into a variety of categories including proteins, glycoproteins, oncofetal antigens, hormones, receptors, genetic markers, and RNA molecules.11
Cancer biomarkers are also classified into prediction, detection, diagnostic, prognostic, and pharmacodynamics biomarkers.20 Prognostic biomarkers are based on the distinguishing features between benign and malignant tumors. Predictive biomarkers (also known as response markers) are used exclusively in assessing the effect of administering a specific drug, thus, allowing clinicians to select a set of chemotherapeutic agents, which will work best for an individual patient. Pharmacodynamic biomarkers are cancer markers utilized in selecting doses of chemotherapeutic agents in a given set of tumor-patient conditions. Diagnostic markers may be present in any stage during cancer development.
HIV-associated Kaposi Sarcoma and its Problems in Diagnosis
Kaposi sarcoma (KS) is an endothelial neoplasia that is found typically in cutaneous lesions, whose development stages entail macules, plaques, and nodules.21 KS is the most common malignancy in HIV patients. HIV-associated Kaposi sarcoma (HIV-KS) is a low-grade vascular tumor associated with human herpesvirus 8 (HHV8)/KS-associated herpes virus infection and is the most aggressive and frequent type of KS.22,23 KS primarily involves the skin but can also involve the viscera.24 Multiple mucocutaneous lesions typically evolve from flat macule (early or patch stage) into plaques (plaque stage) and then nodules (tumor or nodular stage) containing spindle-shaped tumor cells. KS has a variable clinical course, ranging from minimal disease presenting as an incidental finding to a rapidly progressing neoplasm that can result in significant morbidity and mortality, depending on the specific site of involvement.
It poses problems in histologic diagnosis due to its broad morphologic spectrum and mimicry of many benign vasoproliferative lesions and tumors with a prominent spindle component.25 Distinguishing KS from other benign or malignant vascular tumors, as well as other nonvascular spindle cell soft tissue neoplasms, can be challenging.26 Early-stage KS represents a reactive lesion that can either regress or progress. Progression is related to the long-lasting expression of HHV8 latency genes in KS lesions, including latent nuclear antigen-1 (LANA-1),21 cyclin-D1,27,28 and bcl-2.29 HHV8-related induction of the receptor tyrosine kinase c-kit was shown by gene expression profiling in cultured endothelial cells to play a key role in KS tumorigenesis.30,31
Biomarkers Used in HIV-KS Diagnosis/Prognosis
The differential diagnosis of KS may include cutaneous angiosarcoma, spindle cell hemangioma, dermatofibrosarcoma protuberans, vascular transformation of lymph nodes, pilar leiomyoma, stasis dermatitis, pyogenic granuloma, and spindled melanoma among others (Table 1).26 Histologically, all epidemiologic forms of KS are characterized by the progressive proliferation of spindle-shaped cells and are associated with KSHV/HHV8.32 Thus, immunohistochemical detection of HHV8 in fixed tissues would be diagnostically useful, enabling one to differentiate KS from other entities. In latency, HHV8 genes produce numerous proteins that induce or maintain KS lesions, including K12, K13/viral FADD-like interferon converting enzyme inhibitory protein (vFLIP), vCyclin, and the LANA-1 that modulates cellular transcription.33–35
Table 1.
Summary of current biomarkers in HIV-KS.
| BIOMARKER | CHANGES SEEN IN HIV-KS | REFERENCES |
|---|---|---|
| HHV8/LANA-1 | Elevated | 21,22,25,26,29,32 |
| Cyclin D1 | Elevated | 22,27,28 |
| bcl2 | Elevated | 29,33,35,51 |
| c-kit | Elevated | 30,31 |
| k12 | Elevated | 33,34 |
| k13/vFLIP | Elevated | 33,46,47 |
| vCyclin | Elevated | 35,43,45 |
| P53 | Suppressed | 36,37 |
| pRb | Suppressed | 27,33 |
| D2–40 | Elevated | 24,38,40 |
| CD31 | Elevated | 22,24,38,41 |
| CD34 | Elevated | 24,38,41 |
| FLI1 | Elevated | 38 |
| vIL-6 | Elevated | 33,44,52 |
| Tat | Elevated | 49 |
| bFGF | Elevated | 42 |
| TNF-α | Elevated | 42,52 |
| IL-1 | Elevated | 42,51,52 |
| Oncostatin M | Elevated | 42,53,54 |
Abbreviations: HHV8, human herpesvirus 8; LANA-1, latent nuclear antigen-1; bcl2, B-cell lymphoma 2; K13/vFLIP, K13/viral FADD-like interferon converting enzyme inhibitory protein; vCyclin, viral cyclin; pRb, retinoblastoma protein; FLI1, friend leukemia integration-1 transcription factor; vIL-6, viral interleukin-6; TNF-α, tumour necrosis factor-α.
HHV8 LANA-1 is a protein encoded by open reading frame-73 (ORF73) of the virus’ genome. The protein is expressed predominantly during viral latency and appears to play a role in viral integration into the host genome. It has also been shown to interfere in apoptosis via interactions with p53.36 LANA-1 protein may cause dysfunction of cell cycle regulatory checkpoints by degrading p53 and inactivating pRb.37 It has been previously shown that positive immunostaining for HHV8 LANA-1 exhibits high sensitivity and specificity, and it is a reliable and cost-effective method for the diagnosis of KS and is also useful for distinguishing it from the mimickers.21,25,29
Recently, it has been reported that immunohistochemical staining with D2-40, CD31 (a platelet/endothelial cell adhesion molecule, PECAM1), CD34 (a hematopoietic progenitor cell surface protein), and FLI1 (Friend leukemia virus integration 1) is useful for distinguishing cutaneous KS from other diseases.24,38 D2-40 is a novel monoclonal antibody, directed against Mr 40000 O-linked sialoglycoprotein, which reacts with a fixation resistant epitope on the lymphatic endothelium.39 It is considered to be a selective marker of lymphatic endothelium.40 Monoclonal antibodies directed against CD31 and CD34 are sensitive and specific markers of endothelial differentiation that are expressed by the majority of vascular tumors. It has been previously demonstrated that immunostaining for CD31 and CD34 can be used as an aid in KS diagnosis in routinely processed tissues.41 In a study by Rosado and colleagues, it was reported that CD31, CD34, D2-40, and FLI1 markers demonstrated high sensitivity in both AIDS-related and non-AIDS-related KS as well as in stages of tumor progression.38
A number of inflammatory cytokines, peptide growth factors, HIV encoded Tat protein, and KSHV/HHV8 gene products contribute to KS cell growth and development.42 HHV8 antigens target cell signaling pathways and deregulate apoptosis and immune response through vCyclin, vFLIP, bcl-2 oncogene, viral interferon regulating factor, and vIL-6.33,43–47 The alterations of immune cells (lymphocytes, monocytes, histiocytes, and dendritic cells) have been suggested to play a role in the neoplastic process.48 Immune activation can cooperate with some growth factors and HIV-1 Tat protein in the development and progression of KS.49 HIV-KS cells have been shown to produce angiogenic growth factors and cytokines such as fibroblast growth factors (FGFs), tumor necrosis factor-α (TNF-α), interleukin 1 (IL-1), IL-6, Tat, and oncostatin M, and express high affinity receptors for several cytokines.50,51 Elevated levels of IL-1, IL-6, and TNF-α have been reported in patients with HIV-KS.52
Oncostatin M, a cytokine produced by microphages and activated T-lymphocytes, has been shown to be a mitogen for HIV-KS derived spindle cells.53 Oncostatin M appears to be a major cytokine responsible for maintaining the long-term growth of HIV-KS in cell cultures.54 In addition, inflammatory cytokines induce the production of a potent autocrine growth factor for spindle cells known as basic fibroblast growth factor (bFGF). The autologous production of bFGF is an important stage in KS tumorigenesis since antisense bFGF or anti bFGF antibodies interfere in KS cell growth in tissue culture.42 It has been shown that oncostatin M, IL-1, and TNF-α induce KS cell growth by inducing the expression of various bFGF isoforms.
HIV-associated NHL and its Problems in Diagnosis
NHL refers to a heterogeneous group of hematopoietic malignancies originating in the lymphocytes.55–57 The majority of NHL cases (85–90%) arises from B-cell progenitors and develops into the various entities largely grouped into low, intermediate, and high-grade NHL based on the treated natural history and survival patterns.58 NHL comprises many subtypes, each with distinct epidemiology, etiology, and features (ie, morphology, immunophenotype, and clinical manifestations).59,60 Epstein–Barr virus (EBV) has been implicated in the development of many NHL subtypes in HIV-infected individuals.61 NHL is the second most common malignancy in HIV-infected patients, with diffuse large B-cell lymphoma (DLBCL) as the most common subtype of HIV-associated NHL (HIV-NHL) followed by Burkitt’s lymphoma (BL).62
DLBCLs are heterogeneous diseases that differ in nature of the genetic abnormalities, morphologic appearance, clinical features, and patients respond differently to treatment and vary in prognosis.63,64 Most DLBCLs are thought to arise from normal antigen exposed B-cells that have migrated to or through germinal centers.65 Gene expression profiling has identified two broad subgroups: those of germinal center origin, known as germinal centre B-cell like (GCB) lymphomas (typically CD10+ and BCL6+); and those arising from cells resembling activated B-cells (non-GCB) (typically IRF4/MUM-1+/− and CD13+).56,66 It has been shown that patients with GCB DLBCL have a better progression free and overall survival than those with non-GCB DLBCL, irrespective of the international prognostic index (IPI) score.67–71 Therefore, the subclassifications of DLBCL into GCB and non-GCB may serve as important predictive prognostic factors. BL is an aggressive form of NHL derived from germinal center B-cells.72 HIV-associated BL is characterized by cMYC translocations and overexpression;73 however, EBV infection is not necessarily a precursor to transformation.74 NHL is a very complex malignancy consisting of several types that are also divided into subclasses that differ in treatment response and prognosis. This may pose problems in the initial diagnosis of NHL. Biomarkers are necessary in the initial evaluation of the patients with newly diagnosed NHL, which must establish the precise histologic subtype, the extent, and site of disease (localized or advanced, nodal or extranodal). This is important in the determination of treatment approach and predicting the response to chemotherapy.
Biomarkers Used in HIV-NHL Diagnosis/Prognosis
The first step in the diagnosis of NHL is to obtain good quality and adequate sample of tissue by excisional biopsy of an affected lymph node or other mass lesion for assessment of cellular morphology and nodal architecture (Table 2).75–77 After the initial tissue biopsy provides a diagnosis of NHL, the following laboratory tests are performed: complete blood count, white blood cell differential, platelet count, and examination of the peripheral smear for the presence of atypical cells, suggesting peripheral blood and bone marrow involvement; biochemical tests including blood urea nitrogen (BUN), creatinine, alkaline phosphatase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase (LDH), and albumin; serum calcium, electrolytes, and uric acid; serum protein electrophoresis; HIV, hepatitis B, and C serology; and beta-2 microglobulin levels (in patients with indolent lymphomas).78
Table 2.
Summary of current biomarkers in HIV-NHL.
| BIOMARKER | CHANGES SEEN IN HIV-NHL | REFERENCES |
|---|---|---|
| LDH | Elevated | 101,105,106 |
| Ki-67/MIB-1 | Elevated | 61,79,80 |
| CD19, CD20, CD22 | Elevated | 63,89,90 |
| CD79a | Elevated | 63,90 |
| PAX-5 | Elevated | 63,81,81 |
| CD10 | Elevated | 61,62 |
| bcl6 | Elevated | 61,62,65,87 |
| MUM-1 | Elevated | 61,62,65 |
| cMYC | Translocation and elevated | 73,86,87 |
| IL-6 | Elevated | 97,99 |
| IL-10 | Elevated | 95,97,99 |
| TNF-α | Elevated | 97,98 |
| CRP | Elevated | 99,101 |
| sCD23, sCD27, sCD30, sCD44 | Elevated | 48,92,99 |
| B2M | Elevated | 105,106 |
| CXCL13 | Elevated | 48,92 |
| EBV DNA | Elevated | 108 |
| FLC | Elevated | 97,100 |
| FOXP1 | Elevated | 83,85 |
Abbreviations: LDH, lactate dehydrogenase; MIB-1, E3 ubiquitin-protein ligase MIB1; PAX-5, paired box protein 5; bcl6, B-cell lymphoma 6; MUM-1, multiple myeloma oncogene 1; IL-6, interleukin 6; IL-10, interleukin 10; TNF-α, tumor necrosis factor-α; CRP, C-reactive protein; sCD23, soluble CD23; sCD44, soluble CD44; B2M, beta-2 microglobulin; CXCL13, C-X-C motif chemokine 13; EBV DNA, Epstein–Barr virus deoxyribonucleic acid; FLC, free immunoglobulin light chains; FOXP1, forkhead box protein P1.
This is followed by pathological evaluations, which include flow cytometry or immunohistochemical staining for immunophenotype.75 For aggressive lymphomas, this includes evaluation of proliferative fraction using Ki-67 or MIB-1 staining as a more aggressive regimen may be indicated for high growth fraction tumors.77 The expression of Ki-67 has been associated with poor outcome and survival in DLBCL patients.79,80 Immunophenotypic expression patterns of DLBCL include positivity for various pan B-cell markers such as CD19, CD20, CD22, CD79a, PAX-5, and demonstration of immunoglobulin surface light chain restriction by flow cytometry in the majority of cases.63 The presence of positive PAX-5 immunostaining has been strongly associated with B-cell differentiation as PAX-5 is a B-cell restricted transcription factor.81,82 Staining for CD10, bcl-6, and MUM-1 are usually routinely performed in order to distinguish GCB from non-GCB DLBCL.
Fork box protein P1 (FOXP1), an essential transcription regulator of B-cell development, has been shown to be overexpressed in non-GCB DLBCL as compared with GCB DLBCL.71,83,84 FOXP1 has also been associated with poor survival and prognosis.85 It is now recognized that FOXP1 may serve as an additional biomarker for distinguishing non-GCB from GCB DLBCL and should be included in the diagnosis/prognosis of DLBCL. In addition, fluorescence in situ hybridization (FISH) analysis for cMYC is performed as translocations involving the cMYC occurs in 10–15% of DLBCL lymphomas and is associated with a worse outcome.86 MYC translocations confers a worse prognosis in patients treated with cyclophosphamide, hydroxydaunorubicin, oncovin, and prednisone (CHOP), and CHOP plus rituximab (R-CHOP) regimens.87 BL expresses a germinal center B-cell phenotype,88 and the immunophenotypic expression include B-cell antigens CD19, CD20, CD22, CD79a, and PAX-5 along with CD10, bcl-6, CD77, Ki-67 or MIB-1 and monotypic surface light chains such as IgM.89 BL also expresses CD43, TCL1, and CD38 but is negative for CD5, CD23, CD44, CD138, CD34, and TdT.90,91
Altered immune mechanisms play a critical role in the pathogenesis of NHL, as evidenced by increased rates of NHL among HIV-positive patients, transplant recipients, and autoimmune disease patients.92,93 A marked increase in B-cell activation is commonly seen in HIV infection, which is driven by the overproduction of B-cell stimulatory cytokines, such as IL-6 and IL-10, as well as by stimulation of B-cells by HIV and other microbial antigens.94 In addition, HIV itself induces the production of inflammatory cytokines that cause B-cell stimulation, proliferation, and activation, and the cell lines derived from HIV-NHL have been found to express cytokines including interleukin 6, 10, and tumor necrosis factor-α.95–98 B-cell activation is characterized by lymphocyte proliferation, class switch recombination (CSR), and somatic hypermutation, all of which are prone to resultant errors in DNA that may lead to lymphomagenesis. B-cell activation leads to the expression of activation induced cytidine deaminase (AICDA), a DNA modifying enzyme that mediates immunoglobulin gene CSR and somatic hypermutation.99
Various factors associated with B-cell activation, including B-cell stimulatory cytokines, as well as soluble serum molecules that are associated with B-cell activation, including serum immunoglobulins (Ig) and Ig components such as free light chains, have been seen to be elevated preceding the appearance of HIV-NHL.99,100 In a nested case–control study by Breen and colleagues, it was shown that serum levels of molecules associated with B-cell activation including IL-6, IL-10, C-reactive protein (CRP), sCD23, sCD27, and sCD30 are elevated for several years preceding the diagnosis of systemic HIV-NHL.99,101 In addition, De Roos and colleagues, in a case–control study within Women’s Health Initiative study cohort of 491 cases and 491 controls, showed that women with high serum levels of soluble sCD23, sCD27, sCD30, sCD44, and CXCL13 biomarkers were at 2.8- to 5.5-fold increased risk of B-NHL.92 Furthermore, this was confirmed by Hussain and colleagues, in a nested case–control study of 3768 women, where it was shown that elevated levels of sCD27, sCD30, CD23, and CXCL13 were associated with subsequent diagnosis of HIV-NHL.48
Factors associated with poor clinical outcome and shorter survival in patients with HIV-NHL include CD4 cell count <100 mm3, advanced stage disease (III or IV), age over 35 years, history of injection drug use, elevated serum LDH (above normal), Eastern Co-operative Oncology Group performance status (ECOG PS) of more than 2, and the involvement of more than 2 extranodal sites.102–106 Matthews and colleagues, in a cohort of 7840 HIV-positive patients, showed that age, nadir CD4 cell count, and no prior cART are significantly associated with the development of systemic NHL.107 In addition, Tedeschi and colleagues showed that low CD4 and CD8 cell count and detectable EBV viremia are three independent prognostic biomarkers that might help in the management of HIV-NHL patients.108 Furthermore, higher HIV viral load accompanied by lower CD4 count have been associated with the development of HIV-NHL.109,110 It has been shown that the risk of HIV-NHL rises substantially in patients with HIV RNA levels greater than 100 000 copies/μL and those with CD4 lymphocyte counts of less than 50 mm3/μL.111
HIV-associated Cervical Cancer and its Problems in Diagnosis
Human papillomavirus (HPV) infection is the most important cause of cervical cancer; however, only 2% of cervical HPV will develop into cervical cancer.112 Cervical cancer is caused by a persistent infection with high-risk human papillomavirus (hrHPV) types, which lead to premalignant precursor lesions known as cervical intraepithelial neoplasia (CIN).113 The most common histologic types of cervical cancer are squamous cell (69%) and adenocarcinoma (25%).114 CIN is characterized by abnormal cellular proliferation, maturation, and nuclear atypia.115 CIN may regress to normal or progress to invasive cervical cancer if left untreated. Approximately, one-third to one half of the cases of CIN I and CIN II regress without treatment. However, the more severe the abnormality of the lesion, the less likely it is to regress. The accurate grading of CIN lesions is important for clinical management of patients, because CIN I and CIN II/III lesions are treated differently and inaccurate grading results in over or under treatment. This emphasizes the need for specific biomarkers to aid objective CIN grading and to achieve more accurate diagnosis.
Biomarkers Used in HIV-associated Cervical Cancer Diagnosis/Prognosis
The detection of HPV DNA in cervical cancer has been proven to be a good diagnostic and risk predictor tool for cervical cancer (Table 3).116 The oncogenic process in cervical cancer is initiated and mediated by the upregulation of HPV E6/E7 oncoproteins, and thus, overexpression of these oncoproteins is a marker for an increased risk of cervical cancer.117–119 The hrHPV subtypes such as 16 and 18 are thought to play a role in malignant transformation of cells by producing E6 and E7 viral regulatory proteins.33 E6 and E7 are involved in cell proliferation and survival. HPV and oncogene E6 and E7 expressions are the most important markers implicated for cervical cancer.20 Some studies suggest that HPV oncogenes E6 and E7 mRNA levels in the uterine cervix may be more specific early indicators of predisposition to carcinogenesis than DNA levels.120
Table 3.
Summary of current biomarkers in HIV-associated cervical cancer.
| BIOMARKER | CHANGES SEEN IN HIV ASSOCIATED CERVICAL CANCER | REFERENCES |
|---|---|---|
| HPV DNA | Elevated | 20,113,118 |
| HPVE6/E7 | Elevated | 116–120 |
| Ki-67 | Elevated | 121,124,125 |
| P16 | Elevated | 113,121,123,124,126 |
| CK17 | Elevated | 113,126–128 |
| MCM | Elevated | 129,130,132 |
| CDC6 | Elevated | 131,132 |
| Ribosomal protein S12 | Elevated | 133 |
| P53 | Elevated | 115,125,134 |
| PCNA | Elevated | 115,135,137 |
| MIB-1 | Elevated | 137 |
| P63 | suppressed | 113,136 |
| CD44 | Elevated | 138,139 |
Abbreviations: HPV DNA, Human papillomavirus deoxyribonucleic acid; P16, cyclin-dependent kinase inhibitor p16; CK17, cytokeratin 17; MCM, mini chromosome maintenance protein; CDC6, cell division cycle protein 6; PCNA, proliferating cell nuclear antigen.
Ki-67 is a well-known cell proliferation marker, useful for confirmation of the diagnosis in ambiguous cases of cervical cancer and CIN grading.121 Ki-67 detects a nuclear antigen that is present only in proliferating cells but absent in resting cells.122 Ki-67 has been found to be more intensely stained in HPV-positive than HPV-negative epithelium. P16INK4A (p16) is a cyclin-dependent kinase (cdk) inhibitor that functions as a specific biomarker used for identification of squamous and glandular dysplastic cervical epithelium with tendency to invasive cervical cancer. It has been suggested that p16 is overexpressed in cervical epithelial cells that are transformed in response to the expression of the hrHPV E7 oncoprotein.113 In a nested study by Carozzi and colleagues, it was shown that p16 overexpression is a marker for CIN2 or worse or for its development within 3 years in HPV-positive women.123 Ki-67 and p16 are complimentary alternative biomarkers for HPV-related neoplasia.124,125 Cytokeratin (CK) 17 is a useful marker for endocervical reserve stem cells, which gives rise to metaplasia and antibody to CK17 is used to differentiate between immature squamous metaplasia and high-grade CIN (CIN III).126 CK17 is specific for reserve cells and immature metaplastic cells; it is not expressed in cervical glandular epithelial cells, squamous cells, or mature squamous metaplastic cells.127,128
Overexpression of mini chromosome maintenance (MCM) proteins is seen in severe dysplastic lesions,129,130 and overexpressed cell division cycle protein 6 (CDC6) is observed in malignant cervical cancer.131,132 The ribosomal protein S12 gene has also been reported as an early molecular diagnostic identifier for the screening of cervical cancer and is a potential target in cancer gene therapy trials.122,133 Tumor suppressor protein p53 is a nuclear phosphoprotein encoded by the p53 gene, whose normal function is to control cell proliferation and apoptosis. Mutations of the p53 gene are frequently found in most invasive cancer, resulting in loss of tumor suppressor functions of wild type p53 and gain of oncogenic functions. Overexpression of p53 has been suggested to be a possible prognostic marker for cervical cancer.134 Madhumati and colleagues showed that proliferating cell nuclear antigen (PCNA) and P53 expression increases with increasing severity of CIN lesions.115 It has been previously shown that upregulation of PCNA is closely associated with hrHPV and progressive CIN, but does not predict outcome in cervical cancer.135 P63, which is a member of the p53 gene family, is expressed in the basal and parabasal cells of mature cervical, vaginal, and vulval squamous epithelium, and also in cervical reserve cells at the transformation zone.113,136 It has been shown that MIB-1 may be a useful marker for identification of low-grade CIN lesion with high proliferative index.137 CD44 is a cell adhesion molecule that has been reported to be correlated with poor prognosis in invasive cervical cancer.122,138,139 The increased serum CD4+ and CD8+ T-cell levels and the presence of large number of natural killer (NK) cells have been associated with a favorable response in patients with cervical cancer treated with neo-adjuvant chemotherapy.
Discussion/Conclusion
Cancer biomarkers offer a great potential for improving the management of cancer at every point from screening and detection, diagnosis, staging, prognosis, and assessment of treatment response.20 Biomarkers offer the hope of early detection as well as tracking cancer progression and recurrence.66 Early detection may help improve survival of HIV-positive cancer patients, as it could help identify HIV-positive individuals at most risk of cancer development distinguish aggressive from indolent malignancies and track disease progression.
Discovery of new biomarkers suitable for clinical application may aid the diagnosis and classification of cancer, which in turn, should lead to better patient stratification.140 Biomarkers do not need to be cancer specific to be useful; certain proteins may help predict response to therapy or aid in the monitoring of disease progression.141 As cancer is increasingly defined by dysregulated pathways, relevant biomarkers may cut across tumor types without showing tissue specificity. Abundance of potential cancer biomarkers have been discovered, however, only few of them have been integrated into clinical practice. This is due to the fact that some of these biomarkers are not highly sensitive and specific for cancer detection. It is well recognized that the road from biomarker discovery, validation, and regulatory approval to the translation into clinical setting could be long and difficult.11
A new era is underway in which cancer detection, diagnosis, and treatment will be guided increasingly by the molecular attributes of the individual patient.142 The future of cancer therapy lies in the use of biomarkers that offer the potential to identify and treat cancer years before it is either visible or symptomatic. In addition, the future of cancer management is expected to be profoundly dependent upon the use of biomarkers that will guide physicians at every step of disease management.143 Cancer biomarkers can be used for the accurate evaluation and management of the disease.
Footnotes
Author Contributions
Conceived and designed the experiments: BF, GS, PB, BR. Analyzed the data: BF, GS, PB, BR. Wrote the first draft of the manuscript: BF. Contributed to the writing of the manuscript: BF, GS, PB, BR. Agree with manuscript results and conclusions: BF, GS, PB, BR. Jointly developed the structure and arguments for the paper: BF, GS, PB, BR. Made critical revisions and approved final version: BF, GS, PB, BR. All authors reviewed and approved of the final manuscript.
ACADEMIC EDITOR: Barbara Guinn, Editor in Chief
FUNDING: This study was supported by, National Research Foundation-German DFG-IRTG project 1522 “HIV/AIDS and associated infectious diseases in Southern Africa” (NRF-IRTG); Columbia University-South Africa (D43) training program for research on HIV associated malignancies; and Harry Crossley foundation research grant. Funding sources have not influenced the content of the paper.
COMPETING INTERESTS: Authors disclose no potential conflicts of interest.
This paper was subject to independent, expert peer review by a minimum of two blind peer reviewers. All editorial decisions were made by the independent academic editor. All authors have provided signed confirmation of their compliance with ethical and legal obligations including (but not limited to) use of any copyrighted material, compliance with ICMJE authorship and competing interests disclosure guidelines and, where applicable, compliance with legal and ethical guidelines on human and animal research participants.
REFERENCES
- 1.Diamandis M, White NM, Yousef GM. Personalized medicine: marking a new epoch in cancer patient management. Mol Cancer Res. 2010;8(9):1175–1187. doi: 10.1158/1541-7786.MCR-10-0264. [DOI] [PubMed] [Google Scholar]
- 2.Sasco AJ, Jaquet A, Boidin E, et al. The challenge of AIDS-related malignancies in sub-Saharan Africa. PLoS One. 2010;5(1):e8621. doi: 10.1371/journal.pone.0008621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Casper C. The increasing burden of HIV-associated malignancies in resource-limited regions. Annu Rev Med. 2011;62:157–170. doi: 10.1146/annurev-med-050409-103711. [DOI] [PubMed] [Google Scholar]
- 4.Ambinder RF, Bhatia K, Martinez-Maza O, Mitsuyasu R. Cancer biomarkers in HIV patients. Curr Opin HIV AIDS. 2010;5(6):531–537. doi: 10.1097/COH.0b013e32833f327e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.McDonald KL. In: Biomarker Discovery, Validation and Clinical Application for Patients Diagnosed with Glioma, Glioma—Exploring Its Biology and Practical Relevance. Ghosh Anirban., editor. InTech; 2011. [Accessed September 1, 2014]. Available at http://www.intechopen.com/books/glioma-exploring-its-biology-andpractical-relevance/biomarker-discovery-validation-and-clinical-application-for-patients-diagnosed-with-glioma. [Google Scholar]
- 6.Verma M. Personalized medicine and cancer. J Pers Med. 2012;2(1):1–14. doi: 10.3390/jpm2010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Park JW, Kerbel RS, Kelloff GJ, et al. Rationale for biomarkers and surrogate end points in mechanism-driven oncology drug development. Clin Cancer Res. 2004;10(11):3885–3896. doi: 10.1158/1078-0432.CCR-03-0785. [DOI] [PubMed] [Google Scholar]
- 8.Biomarkers Definitions Working Group Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69(3):89–95. doi: 10.1067/mcp.2001.113989. [DOI] [PubMed] [Google Scholar]
- 9.Lesko LJ, Atkinson AJ., Jr Use of biomarkers and surrogate endpoints in drug development and regulatory decision making: criteria, validation, strategies. Annu Rev Pharmacol Toxicol. 2001;41:347–366. doi: 10.1146/annurev.pharmtox.41.1.347. [DOI] [PubMed] [Google Scholar]
- 10.Malati T. Tumour markers: an overview. Indian J Clin Biochem. 2007;22(2):17–31. doi: 10.1007/BF02913308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Füzéry AK, Levin J, Chan MM, Chan DW. Translation of proteomic biomarkers into FDA approved cancer diagnostics: issues and challenges. Clin Proteomics. 2013;10(1):13. doi: 10.1186/1559-0275-10-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Srinivas PR, Kramer BS, Srivastava S. Trends in biomarker research for cancer detection. Lancet Oncol. 2001;2(11):698–704. doi: 10.1016/S1470-2045(01)00560-5. [DOI] [PubMed] [Google Scholar]
- 13.Verma M, Manne U. Genetic and epigenetic biomarkers in cancer diagnosis and identifying high risk populations. Crit Rev Oncol Hematol. 2006;60(1):9–18. doi: 10.1016/j.critrevonc.2006.04.002. [DOI] [PubMed] [Google Scholar]
- 14.Miaskowski C, Aouizerat BE. Biomarkers: symptoms, survivorship, and quality of life. Semin Oncol Nurs. 2012;28(2):129–138. doi: 10.1016/j.soncn.2012.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Overdevest JB, Theodorescu D, Lee JK. Utilizing the molecular gateway: the path to personalized cancer management. Clin Chem. 2009;55(4):684–697. doi: 10.1373/clinchem.2008.118554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Frank R, Hargreaves R. Clinical biomarkers in drug discovery and development. Nat Rev Drug Discov. 2003;2(7):566–580. doi: 10.1038/nrd1130. [DOI] [PubMed] [Google Scholar]
- 17.Etzioni R, Urban N, Ramsey S, et al. The case for early detection. Nat Rev Cancer. 2003;3(4):243–252. doi: 10.1038/nrc1041. [DOI] [PubMed] [Google Scholar]
- 18.Ludwig JA, Weinstein JN. Biomarkers in cancer staging, prognosis and treatment selection. Nat Rev Cancer. 2005;5(11):845–856. doi: 10.1038/nrc1739. [DOI] [PubMed] [Google Scholar]
- 19.Heckman-Stoddard BM. Oncology biomarkers: discovery, validation, and clinical use. Semin Oncol Nurs. 2012;28(2):93–98. doi: 10.1016/j.soncn.2012.03.003. [DOI] [PubMed] [Google Scholar]
- 20.Mishra A, Verma M. Cancer biomarkers: are we ready for the prime time? Cancers. 2010;2(1):190–208. doi: 10.3390/cancers2010190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pereira PF, Cuzzi T, Galhardo MC. Immunohistochemical detection of the latent nuclear antigen-1 of the human herpesvirus type 8 to differentiate cutaneous epidemic Kaposi sarcoma and its histological simulators. An Bras Dermatol. 2013;88(2):243–246. doi: 10.1590/S0365-05962013000200010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pantanowitz L, Dezube BJ, Pinkus GS, Tahan SR. Histological characterization of regression in acquired immunodeficiency syndrome-related Kaposi’s sarcoma. J Cutan Pathol. 2004;31(1):26–34. doi: 10.1046/j.0303-6987.2004.0132.x. [DOI] [PubMed] [Google Scholar]
- 23.Groopman JE. AIDS-Related Kaposi Sarcoma: Staging and Treatment. UpToDate. 2013. [Accessed January 6, 2014]. Available at http://www.uptodate.com/contents/aids-related-kaposi-sarcoma-staging-and-treatment?source=search_result&search=Biomarkers+in+Kaposi+Sarcoma&selectedTitle=2~150.
- 24.Nagata N, Igari T, Shimbo T, et al. Diagnostic value of endothelial markers and HHV-8 staining in gastrointestinal Kaposi sarcoma and its difference in endoscopic tumor staging. World J Gastroenterol. 2013;19(23):3608–3614. doi: 10.3748/wjg.v19.i23.3608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cheuk W, Wong KO, Wong CS, Dinkel JE, Ben-Dor D, Chan JK. Immunostaining for human herpesvirus 8 latent nuclear antigen-1 helps distinguish Kaposi sarcoma from its mimickers. Am J Clin Pathol. 2004;121(3):335–342. doi: 10.1309/B8TC-0LBV-H8XY-5MFV. [DOI] [PubMed] [Google Scholar]
- 26.Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17(4):456–460. doi: 10.1038/modpathol.3800061. [DOI] [PubMed] [Google Scholar]
- 27.Horenstein MG, Cesarman E, Wang X, Linkov I, Prieto VG, Louie DC. Cyclin D1 and retinoblastoma protein expression in Kaposi’s sarcoma. J Cutan Pathol. 1997;24(10):585–589. doi: 10.1111/j.1600-0560.1997.tb01088.x. [DOI] [PubMed] [Google Scholar]
- 28.Hong A, Davies S, Stevens G, Lee CS. Cyclin D1 overexpression in AIDS-related and classic Kaposi sarcoma. Appl Immunohistochem Mol Morphol. 2004;12(1):26–30. doi: 10.1097/00129039-200403000-00005. [DOI] [PubMed] [Google Scholar]
- 29.Long E, Ilie M, Hofman V, et al. LANA-1, Bcl-2, Mcl-1 and HIF-1alpha protein expression in HIV-associated Kaposi sarcoma. Virchows Arch. 2009;55(2):159–170. doi: 10.1007/s00428-009-0791-1. [DOI] [PubMed] [Google Scholar]
- 30.Pantanowitz L, Schwartz EJ, Dezube BJ, Kohler S, Dorfman RF, Tahan SR. C-Kit (CD117) expression in AIDS-related, classic, and African endemic Kaposi sarcoma. Appl Immunohistochem Mol Morphol. 2005;13(2):162–166. doi: 10.1097/00129039-200506000-00009. [DOI] [PubMed] [Google Scholar]
- 31.Moses AV, Jarvis MA, Raggo C, et al. Kaposi’s sarcoma-associated herpesvirus-induced upregulation of the c-kit proto-oncogene, as identified by gene expression profiling, is essential for the transformation of endothelial cells. J Virol. 2002;76(16):8383–8399. doi: 10.1128/JVI.76.16.8383-8399.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Schwartz EJ, Dorfman RF, Kohler S. Human herpesvirus-8 latent nuclear antigen-1 expression in endemic Kaposi sarcoma: an immunohistochemical study of 16 cases. Am J Surg Pathol. 2003;27(12):1546–1550. doi: 10.1097/00000478-200312000-00007. [DOI] [PubMed] [Google Scholar]
- 33.Pulitzer M. Molecular diagnosis of infection-related cancers in dermatopathology. Semin Cutan Med Surg. 2012;31(4):247–257. doi: 10.1016/j.sder.2012.10.001. [DOI] [PubMed] [Google Scholar]
- 34.Cai X, Lu S, Zhang Z, Gonzalez CM, Damania B, Cullen BR. Kaposi’s sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells. Proc Natl Acad Sci U S A. 2005;102(15):5570–5575. doi: 10.1073/pnas.0408192102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ojala PM, Tiainen M, Salven P, et al. Kaposi’s sarcoma-associated herpesvirus-encoded v-cyclin triggers apoptosis in cells with high levels of cyclin-dependent kinase 6. Cancer Res. 1999;59(19):4984–4989. [PubMed] [Google Scholar]
- 36.Friborg J, Jr, Kong W, Hottiger MO, Nabel GJ. p53 inhibition by the LANA protein of KSHV protects against cell death. Nature. 1999;402(6764):889–894. doi: 10.1038/47266. [DOI] [PubMed] [Google Scholar]
- 37.Si H, Robertson ES. Kaposi’s sarcoma-associated herpesvirus-encoded latency-associated nuclear antigen induces chromosomal instability through inhibition of p53 function. J Virol. 2006;80(2):697–709. doi: 10.1128/JVI.80.2.697-709.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rosado FG, Itani DM, Coffin CM, Cates JM. Utility of immunohistochemical staining with FLI1, D2-40, CD31, and CD34 in the diagnosis of acquired immunodeficiency syndrome-related and non-acquired immunodeficiency syndrome-related Kaposi sarcoma. Arch Pathol Lab Med. 2012;136(3):301–304. doi: 10.5858/arpa.2011-0213-OA. [DOI] [PubMed] [Google Scholar]
- 39.Arai E, Kuramochi A, Tsuchida T, et al. Usefulness of D2-40 immunohistochemistry for differentiation between kaposiform hemangioendothelioma and tufted angioma. J Cutan Pathol. 2006;33(7):492–497. doi: 10.1111/j.1600-0560.2006.00461.x. [DOI] [PubMed] [Google Scholar]
- 40.Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15(4):434–440. doi: 10.1038/modpathol.3880543. [DOI] [PubMed] [Google Scholar]
- 41.Russell Jones R, Orchard G, Zelger B, Wilson Jones E. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48(11):1011–1016. doi: 10.1136/jcp.48.11.1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Faris M, Ensoli B, Kokot N, Nel AE. Inflammatory cytokines induce the expression of basic fibroblast growth factor (bFGF) isoforms required for the growth of Kaposi’s sarcoma and endothelial cells through the activation of AP-1 response elements in the bFGF promoter. AIDS. 1998;12(1):19–27. doi: 10.1097/00002030-199801000-00003. [DOI] [PubMed] [Google Scholar]
- 43.Kennedy MM, Biddolph S, Lucas SB, et al. Cyclin D1 expression and HHV8 in Kaposi sarcoma. J Clin Pathol. 1999;52(8):569–573. doi: 10.1136/jcp.52.8.569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Aoki Y, Yarchoan R, Wyvill K, Okamoto S, Little RF, Tosato G. Detection of viral interleukin-6 in Kaposi sarcoma-associated herpesvirus-linked disorders. Blood. 2001;97(7):2173–2176. doi: 10.1182/blood.v97.7.2173. [DOI] [PubMed] [Google Scholar]
- 45.Koopal S, Furuhjelm JH, Järviluoma A, et al. Viral oncogene-induced DNA damage response is activated in Kaposi sarcoma tumorigenesis. PLoS Pathog. 2007;3(9):1348–1360. doi: 10.1371/journal.ppat.0030140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sakakibara S, Pise-Masison CA, Brady JN, Tosato G. Gene regulation and functional alterations induced by Kaposi’s sarcoma-associated herpesvirus-encoded ORFK13/vFLIP in endothelial cells. J Virol. 2009;83(5):2140–2153. doi: 10.1128/JVI.01871-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ballon G, Chen K, Perez R, Tam W, Cesarman E. Kaposi sarcoma herpesvirus (KSHV) vFLIP oncoprotein induces B cell transdifferentiation and tumorigenesis in mice. J Clin Invest. 2011;121(3):1141–1153. doi: 10.1172/JCI44417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hussain SK, Hessol NA, Levine AM, et al. Serum biomarkers of immune activation and subsequent risk of non-Hodgkin B-cell lymphoma among HIV-infected women. Cancer Epidemiol Biomarkers Prev. 2013;22(11):2084–2093. doi: 10.1158/1055-9965.EPI-13-0614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen X, Cheng L, Jia X, et al. Human immunodeficiency virus type 1 Tat accelerates Kaposi sarcoma-associated herpesvirus Kaposin A-mediated tumorigenesis of transformed fibroblasts in vitro as well as in nude and immunocompetent mice. Neoplasia. 2009;11(12):1272–1284. doi: 10.1593/neo.09494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tappero JW, Conant MA, Wolfe SF, Berger TG. Kaposi’s sarcoma: epidemiology, pathogenesis, histology, clinical spectrum, staging criteria and therapy. J Am Acad Dermatol. 1993;28(3):371–395. doi: 10.1016/0190-9622(93)70057-z. [DOI] [PubMed] [Google Scholar]
- 51.Simonart T, Van Vooren JP. Interleukin-1 beta increases the BCL-2/BAX ratio in Kaposi’s sarcoma cells. Cytokine. 2002;19(6):259–266. doi: 10.1006/cyto.2002.1964. [DOI] [PubMed] [Google Scholar]
- 52.Guo WX, Antakly T, Cadotte M, et al. Expression and cytokine regulation of glucocorticoid receptors in Kaposi’s sarcoma. Am J Pathol. 1996;148(6):1999–2008. [PMC free article] [PubMed] [Google Scholar]
- 53.Cai J, Gill PS, Masood R, et al. Oncostatin-M is an autocrine growth factor in Kaposi’s sarcoma. Am J Pathol. 1994;145(1):74–79. [PMC free article] [PubMed] [Google Scholar]
- 54.Amaral MC, Miles S, Kumar G, Nel AE. Oncostatin-M stimulates tyrosine protein phosphorylation in parallel with the activation of p42MAPK/ERK-2 in Kaposi’s cells. Evidence that this pathway is important in Kaposi cell growth. J Clin Invest. 1993;92(2):848–857. doi: 10.1172/JCI116659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.American Cancer Society Cancer Facts and Figures 2010. 2010. [Accessed June 11, 2013]. Available at http://www.cancer.org/acs/groups/content/@nho/documents/document/acspc-024113.pdf.
- 56.Shankland KR, Armitage JO, Hancock BW. Non-Hodgkin lymphoma. Lancet. 2012;380(9844):848–857. doi: 10.1016/S0140-6736(12)60605-9. [DOI] [PubMed] [Google Scholar]
- 57.Pörtner LM, Schönberg K, Hejazi M, et al. T and NK cells of B cell NHL patients exert cytotoxicity against lymphoma cells following binding of bispecific tetravalent antibody CD19 × CD3 or CD19 × CD16. Cancer Immunol Immunother. 2012;61(10):1869–1875. doi: 10.1007/s00262-012-1339-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chan JK. The new World Health Organization classification of lymphomas: the past, the present and the future. Hematol Oncol. 2001;19(4):129–150. doi: 10.1002/hon.660. [DOI] [PubMed] [Google Scholar]
- 59.BioOncology . Non-Hodgkin’s Lymphoma: A Histopathologic and Prognostic Evaluation. Gentech; USA: 2010. [Accessed November 11, 2013]. Available at http://www.biooncology.com/research-education/bcell/downloads/GA10000083900_NHL_Primer.pdf. [Google Scholar]
- 60.Emmanuel B, Anderson WF. Non-Hodgkin lymphoma in early life. J Natl Cancer Inst. 2012;104(12):888–889. doi: 10.1093/jnci/djs252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chao C, Silverberg MJ, Martínez-Maza O, et al. Epstein-Barr virus infection and expression of B-cell oncogenic markers in HIV-related diffuse large B-cell lymphoma. Clin Cancer Res. 2012;18(17):4702–4712. doi: 10.1158/1078-0432.CCR-11-3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Barreto L, Azambuja D, Morais JC. Expression of immunohistochemical markers in patients with AIDS-related lymphoma. Braz J Infect Dis. 2012;16(1):74–77. doi: 10.1016/s1413-8670(12)70278-7. [DOI] [PubMed] [Google Scholar]
- 63.Sangle NA, Agarwal AM, Smock KJ, et al. Diffuse large B-cell lymphoma with aberrant expression of the T-cell antigens CD2 and CD7. Appl Immunohistochem Mol Morphol. 2011;19(6):579–583. doi: 10.1097/PAI.0b013e318221c672. [DOI] [PubMed] [Google Scholar]
- 64.Kim MK, Bae SH, Bae YK, et al. Biological characterization of nodal versus extranodal presentation of diffuse large B-cell lymphoma using immunohistochemistry. Clin Lymphoma Myeloma Leuk. 2011;11(5):403–408. doi: 10.1016/j.clml.2011.05.037. [DOI] [PubMed] [Google Scholar]
- 65.De Mello CA, De Andrade VP, De Lima VC, Carvalho AL, Soares FA. Prognostic impact of MUM1 expression by immunohistochemistry on primary mediastinal large B-cell lymphoma. Leuk Lymphoma. 2011;52(8):1495–1503. doi: 10.3109/10428194.2011.573032. [DOI] [PubMed] [Google Scholar]
- 66.Dave H, Learn C, Lieberman R. Biomarkers: Recent Advances in Their Application to the Treatment of Hematologic Malignancies. Quintiles; 2013. [Accessed May 1, 2014]. Available at http://www.quintiles.com/library/white-papers/biomarkers-recent-advances-in-their-application-to-the-treatment-of-hematologic-malignancies.pdf. [Google Scholar]
- 67.Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503–511. doi: 10.1038/35000501. [DOI] [PubMed] [Google Scholar]
- 68.Nyman H, Adde M, Karjalainen-Lindsberg ML, et al. Prognostic impact of immunohistochemically defined germinal center phenotype in diffuse large B-cell lymphoma patients treated with immunochemotherapy. Blood. 2007;109(11):4930–4935. doi: 10.1182/blood-2006-09-047068. [DOI] [PubMed] [Google Scholar]
- 69.Zinzani PL, Dirnhofer S, Sabattini E, et al. Identification of outcome predictors in diffuse large B-cell lymphoma. Immunohistochemical profiling of homogeneously treated de novo tumors with nodal presentation on tissue micro-arrays. Haematologica. 2005;90(3):341–347. [PubMed] [Google Scholar]
- 70.Habara T, Sato Y, Takata K, et al. Germinal center B-cell-like versus non-germinal center B-cell-like as important prognostic factor for localized nodal DLBCL. J Clin Exp Hematop. 2012;52(2):91–99. doi: 10.3960/jslrt.52.91. [DOI] [PubMed] [Google Scholar]
- 71.Visco C, Li Y, Xu-Monette ZY, et al. Comprehensive gene expression profiling and immunohistochemical studies support application of immunophenotypic algorithm for molecular subtype classification in diffuse large B-cell lymphoma: a report from the International DLBCL Rituximab-CHOP Consortium Program Study. Leukemia. 2012;26(9):2103–2113. doi: 10.1038/leu.2012.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Schmitz R, Young RM, Ceribelli M, et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature. 2012;490(7418):116–120. doi: 10.1038/nature11378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mead GM, Barrans SL, Qian W, et al. A prospective clinicopathologic study of dose-modified CODOX-M/IVAC in patients with sporadic Burkitt lymphoma defined using cytogenetic and immunophenotypic criteria (MRC/NCRI LY10 trial) Blood. 2008;112(6):2248–2260. doi: 10.1182/blood-2008-03-145128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Levine AM. Challenges in the management of Burkitt’s lymphoma. Clin Lymphoma. 2002;3(suppl 1):S19–S25. doi: 10.3816/clm.2002.s.011. [DOI] [PubMed] [Google Scholar]
- 75.Armitage JO. How I treat patients with diffuse large B-cell lymphoma. Blood. 2007;110(1):29–36. doi: 10.1182/blood-2007-01-041871. [DOI] [PubMed] [Google Scholar]
- 76.Steinfort DP, Conron M, Tsui A, et al. Endobronchial ultrasound-guided transbronchial needle aspiration for the evaluation of suspected lymphoma. J Thorac Oncol. 2010;5(6):804–809. doi: 10.1097/jto.0b013e3181d873be. [DOI] [PubMed] [Google Scholar]
- 77.Kaplan LD. HIV-associated lymphoma. Best Pract Res Clin Haematol. 2012;25(1):101–117. doi: 10.1016/j.beha.2012.01.001. [DOI] [PubMed] [Google Scholar]
- 78.Freedman AS, Friedberg JW. Evaluation and Staging of Non-Hodgkin Lymphoma. UpToDate. 2013. [Accessed June 1, 2014]. Available at http://www.uptodate.com/contents/evaluation-and-staging-of-non-hodgkin-lymphoma?source=search_result&search=non+hodgkins+lymphoma&selectedTitle=5~150.
- 79.Hasselblom S, Ridell B, Sigurdardottir M, Hansson U, Nilsson-Ehle H, Andersson PO. Low rather than high Ki-67 protein expression is an adverse prognostic factor in diffuse large B-cell lymphoma. Leuk Lymphoma. 2008;49(8):1501–1509. doi: 10.1080/10428190802140055. [DOI] [PubMed] [Google Scholar]
- 80.Li ZM, Huang JJ, Xia Y, et al. High Ki-67 expression in diffuse large B-cell lymphoma patients with non-germinal center subtype indicates limited survival benefit from R-CHOP therapy. Eur J Haematol. 2012;88(6):510–517. doi: 10.1111/j.1600-0609.2012.01778.x. [DOI] [PubMed] [Google Scholar]
- 81.Dong HY, Browne P, Liu Z, Gangi M. PAX-5 is invariably expressed in B-cell lymphomas without plasma cell differentiation. Histopathology. 2008;53(3):278–287. doi: 10.1111/j.1365-2559.2008.03091.x. [DOI] [PubMed] [Google Scholar]
- 82.Desouki MM, Post GR, Cherry D, Lazarchick J. PAX-5: a valuable immunohistochemical marker in the differential diagnosis of lymphoid neoplasms. Clin Med Res. 2010;8(2):84–88. doi: 10.3121/cmr.2010.891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yu B, Zhou X, Li B, Xiao X, Yan S, Shi D. FOXP1 expression and its clinicopathologic significance in nodal and extranodal diffuse large B-cell lymphoma. Ann Hematol. 2011;90(6):701–708. doi: 10.1007/s00277-010-1124-9. [DOI] [PubMed] [Google Scholar]
- 84.Sagardoy A, Martinez-Ferrandis JI, Roa S, et al. Downregulation of FOXP1 is required during germinal center B-cell function. Blood. 2013;121(21):4311–4320. doi: 10.1182/blood-2012-10-462846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Hu CR, Wang JH, Wang R, Sun Q, Chen LB. Both FOXP1 and p65 expression are adverse risk factors in diffuse large B-cell lymphoma: a retrospective study in China. Acta Histochem. 2013;115(2):137–143. doi: 10.1016/j.acthis.2012.06.001. [DOI] [PubMed] [Google Scholar]
- 86.Ladanyi M, Offit K, Jhanwar SC, Filippa DA, Chaganti RS. MYC rearrangement and translocations involving band 8q24 in diffuse large cell lymphomas. Blood. 1991;77(5):1057–1063. [PubMed] [Google Scholar]
- 87.Horn H, Ziepert M, Becher C, et al. MYC status in concert with BCL2 and BCL6 expression predicts outcome in diffuse large B-cell lymphoma. Blood. 2013;121(12):2253–2263. doi: 10.1182/blood-2012-06-435842. [DOI] [PubMed] [Google Scholar]
- 88.Whitten J, Arcila ME, Teruya-Feldstein J. Burkitt lymphoma. Pathol Case Rev. 2012;17:79–83. [Google Scholar]
- 89.Miles RR, Arnold S, Cairo MS. Risk factors and treatment of childhood and adolescent Burkitt lymphoma/leukaemia. Br J Haematol. 2012;156(6):730–743. doi: 10.1111/j.1365-2141.2011.09024.x. [DOI] [PubMed] [Google Scholar]
- 90.de Leval L, Hasserjian RP. Diffuse large B-cell lymphomas and Burkitt lymphoma. Hematol Oncol Clin North Am. 2009;23(4):791–827. doi: 10.1016/j.hoc.2009.04.004. [DOI] [PubMed] [Google Scholar]
- 91.Linch DC. Burkitt lymphoma in adults. Br J Haematol. 2012;156(6):693–703. doi: 10.1111/j.1365-2141.2011.08877.x. [DOI] [PubMed] [Google Scholar]
- 92.De Roos AJ, Mirick DK, Edlefsen KL, et al. Markers of B-cell activation in relation to risk of non-Hodgkin lymphoma. Cancer Res. 2012;72(18):4733–4743. doi: 10.1158/0008-5472.CAN-12-1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Mellgren K, Hedegaard CJ, Schmiegelow K, Müller K. Plasma cytokine profiles at diagnosis in pediatric patients with non-Hodgkin lymphoma. J Pediatr Hematol Oncol. 2012;34(4):271–275. doi: 10.1097/MPH.0b013e3182431e02. [DOI] [PubMed] [Google Scholar]
- 94.Vendrame E, Martínez-Maza O. Assessment of pre-diagnosis biomarkers of immune activation and inflammation: insights on the etiology of lymphoma. J Proteome Res. 2011;10(1):113–119. doi: 10.1021/pr100729z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Masood R, Zhang Y, Bond MW, et al. Interleukin-10 is an autocrine growth factor for acquired immunodeficiency syndrome-related B-cell lymphoma. Blood. 1995;85(12):3423–3430. [PubMed] [Google Scholar]
- 96.Powles T, Matthews G, Bower M. AIDS related systemic non-Hodgkin’s lymphoma. Sex Transm Infect. 2000;76(5):335–341. doi: 10.1136/sti.76.5.335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Vendrame E, Hussain SK, Breen EC, et al. Serum levels of cytokines and biomarkers for inflammation and immune activation, and HIV-associated non-Hodgkin B-cell lymphoma risk. Cancer Epidemiol Biomarkers Prev. 2014;23(2):343–349. doi: 10.1158/1055-9965.EPI-13-0714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Nakayama S, Yokote T, Hirata Y, et al. TNF-α expression in tumor cells as a novel prognostic marker for diffuse large B-cell lymphoma, not otherwise specified. Am J Surg Pathol. 2014;38(2):228–234. doi: 10.1097/PAS.0000000000000094. [DOI] [PubMed] [Google Scholar]
- 99.Breen EC, Hussain SK, Magpantay L, et al. B-cell stimulatory cytokines and markers of immune activation are elevated several years prior to the diagnosis of systemic AIDS-associated non-Hodgkin B-cell lymphoma. Cancer Epidemiol Biomarkers Prev. 2011;20(7):1303–1314. doi: 10.1158/1055-9965.EPI-11-0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Landgren O, Goedert JJ, Rabkin CS, et al. Circulating serum free light chains as predictive markers of AIDS-related lymphoma. J Clin Oncol. 2010;28(5):773–779. doi: 10.1200/JCO.2009.25.1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Suzuki K, Terui Y, Nishimura N, et al. Prognostic value of C-reactive protein, lactase dehydrogenase and anemia in recurrent or refractory aggressive lymphoma. Jpn J Clin Oncol. 2013;43(1):37–44. doi: 10.1093/jjco/hys194. [DOI] [PubMed] [Google Scholar]
- 102.Ratner L, Lee J, Tang S, et al. Chemotherapy for human immunodeficiency virus-associated non-Hodgkin’s lymphoma in combination with highly active antiretroviral therapy. J Clin Oncol. 2001;19(8):2171–2178. doi: 10.1200/JCO.2001.19.8.2171. [DOI] [PubMed] [Google Scholar]
- 103.Ansell SM, Armitage J. Non-Hodgkin lymphoma: diagnosis and treatment. Mayo Clin Proc. 2005;80(8):1087–1097. doi: 10.4065/80.8.1087. [DOI] [PubMed] [Google Scholar]
- 104.Levine AM. AIDS-related lymphoma. Semin Oncol Nurs. 2006;22(2):80–89. doi: 10.1016/j.soncn.2006.01.004. [DOI] [PubMed] [Google Scholar]
- 105.Milanovic N, Matkovic S, Ristic D, Jelic S, Petrovic M. Significance of tumor burden, vascular endothelial growth factor, lactate dehydrogenase and beta-2 microglobulin serum levels in advanced diffuse large B cell lymphoma. J BUON. 2012;17(3):497–501. [PubMed] [Google Scholar]
- 106.Bairey O, Bar-Natan M, Shpilberg O. Early death in patients diagnosed with non-Hodgkin’s lymphoma. Ann Hematol. 2013;92(3):345–350. doi: 10.1007/s00277-012-1623-y. [DOI] [PubMed] [Google Scholar]
- 107.Matthews GV, Bower M, Mandalia S, Powles T, Nelson MR, Gazzard BG. Changes in acquired immunodeficiency syndrome-related lymphoma since the introduction of highly active antiretroviral therapy. Blood. 2000;96(8):2730–2734. [PubMed] [Google Scholar]
- 108.Tedeschi R, Bortolin MT, Bidoli E, et al. Assessment of immunovirological features in HIV related non-Hodgkin lymphoma patients and their impact on outcome. J Clin Virol. 2012;53(4):297–301. doi: 10.1016/j.jcv.2011.12.021. [DOI] [PubMed] [Google Scholar]
- 109.Guiguet M, Boué F, Cadranel J, et al. Effect of immunodeficiency, HIV viral load, and antiretroviral therapy on the risk of individual malignancies (FHDH-ANRS CO4): a prospective cohort study. Lancet Oncol. 2009;10(12):1152–1159. doi: 10.1016/S1470-2045(09)70282-7. [DOI] [PubMed] [Google Scholar]
- 110.Engels EA, Pfeiffer RM, Landgren O, Moore RD. Immunologic and virologic predictors of AIDS-related non-Hodgkin lymphoma in the highly active antiretroviral therapy era. J Acquir Immune Defic Syndr. 2010;54(1):78–84. doi: 10.1097/01.qai.0000371677.48743.8d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zoufaly A, Stellbrink HJ, Heiden MA, et al. Cumulative HIV viremia during highly active antiretroviral therapy is a strong predictor of AIDS-related lymphoma. J Infect Dis. 2009;200(1):79–87. doi: 10.1086/599313. [DOI] [PubMed] [Google Scholar]
- 112.Izadi-Mood N, Sarmadi S, Eftekhar Z, Jahanteegh HA, Sanii S. Immunohistochemical expression of p16 and HPV L1 capsid proteins as predictive markers in cervical lesions. Arch Gynecol Obstet. 2013 doi: 10.1007/s00404-013-3124-1. [DOI] [PubMed] [Google Scholar]
- 113.Selvi K, Badhe BA, Papa D, Nachiappa Ganesh R. Role of p16, CK17, p63, and human papillomavirus in diagnosis of cervical intraepithelial neoplasia and distinction from its mimics. Int J Surg Pathol. 2013 doi: 10.1177/1066896913496147. [DOI] [PubMed] [Google Scholar]
- 114.Frumovitz M. Invasive Cervical Cancer: Epidemiology, Risk Factors, Clinical Manifestations, and Diagnosis. UpToDate. 2013. [Accessed December 11, 2013]. Available at http://www.uptodate.com/contents/invasive-cervical-cancer-epidemiology-risk-factors-clinical-manifestations-and-diagnosis?source=search_result&search=cervical+cancer&selectedTitle=1~150.
- 115.Madhumati G, Kavita S, Anju M, Uma S, Raj M. Immunohistochemical expression of cell proliferating nuclear antigen (PCNA) and p53 protein in cervical cancer. J Obstet Gynaecol India. 2012;62(5):557–561. doi: 10.1007/s13224-012-0180-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Campbell LM, Pitta DR, De Assis AM, Derchain SF, Campos EA, Sarian LO. Retrieval of HPV oncogenes E6 and E7 mRNA from cervical specimens using a manual open technology protocol. Springerplus. 2013;2:473. doi: 10.1186/2193-1801-2-473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ratnam S, Coutlee F, Fontaine D, et al. Aptima HPV E6/E7 mRNA test is as sensitive as Hybrid Capture 2 Assay but more specific at detecting cervical precancer and cancer. J Clin Microbiol. 2011;49(2):557–564. doi: 10.1128/JCM.02147-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Roncaglia MT, Fregnani JH, Tacla M, et al. Characterization of p16 and E6 HPV-related proteins in uterine cervix high-grade lesions of patients treated by conization with large loop excision. Oncol Lett. 2013;6(1):63–68. doi: 10.3892/ol.2013.1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Tagle DK, Sotelo DH, Illades-Aguiar B, et al. Expression of E6, p53 and p21 proteins and physical state of HPV16 in cervical cytologies with and without low grade lesions. Int J Clin Exp Med. 2014;7(1):186–193. [PMC free article] [PubMed] [Google Scholar]
- 120.Benevolo M, Vocaturo A, Caraceni D, et al. Sensitivity, specificity, and clinical value of human papillomavirus (HPV) E6/E7 mRNA assay as a triage test for cervical cytology and HPV DNA test. J Clin Microbiol. 2011;49(7):2643–2650. doi: 10.1128/JCM.02570-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sari Aslani F, Safaei A, Pourjabali M, Momtahan M. Evaluation of Ki67, p16 and CK17 markers in differentiating cervical intraepithelial neoplasia and benign lesions. Iran J Med Sci. 2013;38(1):15–21. [PMC free article] [PubMed] [Google Scholar]
- 122.Lindström A. Prognostic Factors for Squamous Cell Cervical Cancer: Tumor Markers, Hormones, Smoking, and S-Phase Fraction. Umeå; 2010. [Accessed June 1, 2014]. Available at http://www.diva-portal.org/smash/get/diva2:318860/FULLTEXT01.pdf. [Google Scholar]
- 123.Carozzi F, Gillio-Tos A, Confortini M, et al. Risk of high-grade cervical intraepithelial neoplasia during follow-up in HPV-positive women according to baseline p16-INK4A results: a prospective analysis of a nested substudy of the NTCC randomised controlled trial. Lancet Oncol. 2013;14(2):168–176. doi: 10.1016/S1470-2045(12)70529-6. [DOI] [PubMed] [Google Scholar]
- 124.Iaconis L, Hyjek E, Ellenson LH, Pirog EC. p16 and Ki-67 immunostaining in atypical immature squamous metaplasia of the uterine cervix: correlation with human papillomavirus detection. Arch Pathol Lab Med. 2007;131(9):1343–1349. doi: 10.5858/2007-131-1343-PAKIIA. [DOI] [PubMed] [Google Scholar]
- 125.Portari EA, Russomano FB, de Camargo MJ, et al. Immunohistochemical expression of cyclin D1, p16Ink4a, p21WAF1, and Ki-67 correlates with the severity of cervical neoplasia. Int J Gynecol Pathol. 2013;32(5):501–508. doi: 10.1097/PGP.0b013e31826f5cf6. [DOI] [PubMed] [Google Scholar]
- 126.Regauer S, Reich O. CK17 and p16 expression patterns distinguish (atypical) immature squamous metaplasia from high-grade cervical intraepithelial neoplasia (CIN III) Histopathology. 2007;50(5):629–635. doi: 10.1111/j.1365-2559.2007.02652.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Martens JE, Arends J, Van der Linden PJ, De Boer BA, Helmerhorst TJ. Cytokeratin 17 and p63 are markers of the HPV target cell, the cervical stem cell. Anticancer Res. 2004;2(2B):771–775. [PubMed] [Google Scholar]
- 128.Ikeda K, Tate G, Suzuki T, Mitsuya T. Coordinate expression of cytokeratin 8 and cytokeratin 17 immunohistochemical staining in cervical intraepithelial neoplasia and cervical squamous cell carcinoma: an immunohistochemical analysis and review of the literature. Gynecol Oncol. 2008;108(3):598–602. doi: 10.1016/j.ygyno.2007.11.042. [DOI] [PubMed] [Google Scholar]
- 129.Ishimi Y, Okayasu I, Kato C, et al. Enhanced expression of MCM proteins in cancer cells derived from uterine cervix. Eur J Biochem. 2003;270(6):1089–1101. doi: 10.1046/j.1432-1033.2003.03440.x. [DOI] [PubMed] [Google Scholar]
- 130.Das M, Prasad SB, Yadav SS, et al. Over expression of minichromosome maintenance genes is clinically correlated to cervical carcinogenesis. PLoS One. 2013;8(7):e69607. doi: 10.1371/journal.pone.0069607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Bonds L, Baker P, Gup C, Shroyer KR. Immunohistochemical localization of cdc6 in squamous and glandular neoplasia of the uterine cervix. Arch Pathol Lab Med. 2002;126(10):1164–1168. doi: 10.5858/2002-126-1164-ILOCIS. [DOI] [PubMed] [Google Scholar]
- 132.Murphy N, Ring M, Heffron CC, et al. Quantitation of CDC6 and MCM5 mRNA in cervical intraepithelial neoplasia and invasive squamous cell carcinoma of the cervix. Mod Pathol. 2005;18(6):844–849. doi: 10.1038/modpathol.3800361. [DOI] [PubMed] [Google Scholar]
- 133.Cheng Q, Lau WM, Chew SH, Ho TH, Tay SK, Hui KM. Identification of molecular markers for the early detection of human squamous cell carcinoma of the uterine cervix. Br J Cancer. 2002;86(2):274–281. doi: 10.1038/sj.bjc.6600038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Romus I, Triningsih FE, Mangunsudirdjo S, Harijadi A. Clinicopathology significance of p53 and p63 expression in Indonesian cervical squamous cell carcinomas. Asian Pac J Cancer Prev. 2013;14(12):7737–7741. doi: 10.7314/apjcp.2013.14.12.7737. [DOI] [PubMed] [Google Scholar]
- 135.Branca M, Ciotti M, Giorgi C, et al. Up-regulation of proliferating cell nuclear antigen (PCNA) is closely associated with high-risk human papillomavirus (HPV) and progression of cervical intraepithelial neoplasia (CIN), but does not predict disease outcome in cervical cancer. Eur J Obstet Gynecol Reprod Biol. 2007;130(2):223–231. doi: 10.1016/j.ejogrb.2006.10.007. [DOI] [PubMed] [Google Scholar]
- 136.Zhou Y, Xu Q, Ling B, Xiao W, Liu P. Reduced expression of ΔNp63α in cervical squamous cell carcinoma. Clin Invest Med. 2011;34(3):E184–E191. doi: 10.25011/cim.v34i3.15192. [DOI] [PubMed] [Google Scholar]
- 137.Goel MM, Mehrotra A. Immunohistochemical expression of MIB-1 and PCNA in precancerous and cancerous lesions of uterine cervix. Indian J Cancer. 2013;50(3):200–205. doi: 10.4103/0019-509X.118729. [DOI] [PubMed] [Google Scholar]
- 138.Speiser P, Wanner C, Tempfer C, et al. CD44 is an independent prognostic factor in early-stage cervical cancer. Int J Cancer. 1997;74(2):185–188. doi: 10.1002/(sici)1097-0215(19970422)74:2<185::aid-ijc8>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
- 139.Shimabukuro K, Toyama-Sorimachi N, Ozaki Y, et al. The expression patterns of standard and variant CD44 molecules in normal uterine cervix and cervical cancer. Gynecol Oncol. 1997;64(1):26–34. doi: 10.1006/gyno.1996.4530. [DOI] [PubMed] [Google Scholar]
- 140.Dokmanovic L. Biomarkers in childhood non-Hodgkin’s lymphomas. Biomark Med. 2013;7(5):791–801. doi: 10.2217/bmm.13.84. [DOI] [PubMed] [Google Scholar]
- 141.Hanash SM, Pitteri SJ, Faca VM. Mining the plasma proteome for cancer biomarkers. Nature. 2008;452(7187):571–579. doi: 10.1038/nature06916. [DOI] [PubMed] [Google Scholar]
- 142.Bhatt AN, Mathur R, Farooque A, Verma A, Dwarakanath BS. Cancer biomarkers—current perspectives. Indian J Med Res. 2010;132:129–149. [PubMed] [Google Scholar]
- 143.Chatterjee SK, Zetter BR. Cancer biomarkers: knowing the present and predicting the future. Future Oncol. 2005;1(1):37–50. doi: 10.1517/14796694.1.1.37. [DOI] [PubMed] [Google Scholar]
