Skip to main content

Second malignant neoplasms in lymphomas, secondary lymphomas and lymphomas in metabolic disorders/diseases

Abstract

With inconsistent findings, evidence has been obtained in recent years that metabolic disorders are closely associated with the development of lymphomas. Studies and multiple analyses have been published also indicating that some solid tumor survivors develop a secondary lymphoma, whereas some lymphoma survivors subsequently develop a second malignant neoplasm (SMN), particularly solid tumors. An interaction between the multiple etiologic factors such as genetic factors and late effects of cancer therapy may play an important role contributing to the carcinogenesis in patients with metabolic diseases or with a primary cancer. In this review, we summarize the current knowledge of the multiple etiologic factors for lymphomagenesis, focusing on the SMN in lymphoma, secondary lymphomas in primary cancers, and the lymphomas associated to metabolic disorders/diseases, which have been received less attention previously. Further, we also review the data of coexistence of lymphomas and hepatocellular carcinoma (HCC) in patients with infection of hepatitis C virus and hepatitis B virus.

Graphical Abstract

Introduction

Based on morphology, immunophenotype, genetic alterations, and clinical features, lymphomas have been categorized into more than 80 distinct entities, principally according to the World Health Organization classification [1]. In fact, lymphomas are a heterogeneous group of hematological malignancies and multifactorial etiopathogenesis has been proposed for the development of lymphomas. Lymphoma therapy is varying greatly, based on type of lymphoma, stage, histopathological features, and patient’s factors such as age and symptoms. The most common treatment includes chemotherapy, radiotherapy, immunotherapy, cellular therapy, and in rare cases, surgery. The published studies indicate that some lymphoma survivors subsequently develop a second malignant neoplasm (SMN), particularly solid tumors, whereas some solid tumor survivors develop a secondary lymphoma. Over the years, the disease-related etiologies have been extrapolated from current literature for those patients with metabolic diseases who may be at high risk for lymphoma development. While appropriate clinical manipulation for cancer survivors and patients with metabolic diseases may prevent or even reverse the lymphomagenetic process, it is really important to gain insight into the lymphomagenesis for those patients but received less attention. In this review article, PubMed was searched from inception to 2021 to discuss the available reports on either lymphoma patients subsequently being diagnosed with SMNs or development of secondary lymphomas from the patients with primary cancers, and the association between lymphomas and metabolic diseases. A summary of studies reported in the literature is presented by lymphoma type and subtype, including classical Hodgkin lymphoma (HL), EBV‐Hodgkin lymphoma, non-Hodgkin lymphoma (NHL) including Diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), mantle cell lymphoma (MCL), primary effusion lymphoma (PEL), marginal zone lymphoma (MZL), and T‐cell lymphoma.

Lymphomas and second malignant neoplasms

Over the past decades, treatment of lymphoma in adults and children has become quite successful because of the improved therapeutic radiation techniques [2, 3] and the introduction of effective combination chemotherapeutic regimens [4, 5]. However, it has been noticed a substantial increase of SMN in lymphoma survivors with a longer life span. As childhood cancer survivors have a longer life expectancy than adult cancer survivors, SMN is considered as one of the most devastating late effects of childhood cancers which call attention world-widely because the occurrence of SMNs in childhood cancer survivors shortens their life expectancy and influences their quality of life [6,7,8,9,10]. In a cohort of children and adolescents being treated between 1960 and 1989 for Hodgkin’s disease, cumulative percentage of patients who developed a SMN after diagnosis was 3.82% ± 1.53% at 10 years; 8.02% ± 2.35% at 15 years; 12.71% ± 3.20% at 20 years; 16.11% ± 3.88% at 25 years; and 26.27% ± 6.75% at 30 years, respectively [11]. Recently, a larger cohort of children and adolescents with SMNs after childhood NHL therapy (n = 189) was reported, with the details of 5 major categories of SMNs (myeloid SMNs, lymphoid SMNs, carcinomas, central nervous system (CNS) tumors, “other” SMNs) as well as the associated NHL subtypes [12]. In this study, a higher risk for the patients with lymphoblastic lymphomas to develop an SMN is suggested, while the patients with lymphoid SMNs, carcinomas, and “other” SMNs showed a better outcome than the patients with myeloid SMNs and CNS tumors (p < 0.0001) [12]. Besides the childhood cancer survivors, more adults after a diagnosis of cancer are living longer than ever before. Now, the cancer survivors account for around 5% of the US population, from 3 million in 1971 to more than 16.9 million in 2019, and this number is expected to continue growing to reach 22.1 million by 2030 [13]. The increased lifetime could also result in an increased risk of SMNs in adults, most importantly, the patients who developed secondary malignancies often have a poor prognosis. In a cohort of 6171 NHL patients being identified within the tumor registries in Sweden, Ontario, and Iowa and the Netherlands Cancer Institute, SMNs were reported in 541 subjects [observed-to-expected ratio (O/E) = 1.37; 95% CI = 1.26–1.49], with actuarial risk of 21% for developing a SMN at 3–20 years after diagnosis of NHL, compared to the population expected cumulative risk of 15% [14]. In a cohort of 592 patients being diagnosed as NHL at Osaka Medical Center for Cancer and Cardiovascular Diseases, significantly increased risk was found for HCC (O/E = 4.36, 95% CI = 1.99–8.28; O = 9) and non-lymphocytic leukemia (O/E = 26.17, 95% CI = 5.26–76.46; O = 3). The patients who received chemotherapy as the NHL treatment had a significantly increased risk of HCC (O/E = 5.91, 95% CI = 2.70–11.23; O = 9), in which 8 out of 9 patients showed evidence of cirrhosis at the time of HCC diagnosis [15]. In a cohort of 2,456 patients with NHL who were first treated from 1973 to 2000 from centers in the British National Lymphoma Investigation, the relative risks (RRs) were significantly elevated for all malignancies combined (RR = 1.3; 95% CI, 1.1 to 1.6), for leukemia (RR = 8.8; 95% CI, 5.1 to 14.1) and lung cancer (RR = 1.6; 95% CI, 1.1 to 2.3), compared with expectations based on general population cancer rates [16]. In a larger cohort of 109,451 NHL first primary cases, a significant (P < 0.001) increase was observed for 18 various cancers, including lip, tongue, oropharynx, stomach, small intestine, colon, liver, nose and nasal cavity, lung, soft tissue, skin melanoma and nonmelanoma skin cancer, bladder, kidney, thyroid, Hodgkin’s lymphoma, lymphoid leukaemia and myeloid leukaemia [17]. The overall increase in the risk of a SMN was 47%, while the RR was higher along with the increasing time of follow-up, being 1.37 (95% CI 1.32–1.43) in the 1–4-year follow-up period, and 1.67 (95% CI 1.59–1.76) after 10 years or more [17]. In 2015, a cohort study of 2548 Hodgkin’s lymphoma patients from the German, Austrian, and Swiss with follow up over 30 years reported that the cumulative incidence of SMN at 20, 25, and 30 years was 7%, 11.2%, and 18.7%, respectively [18]. For all types of SMN, the standardized incidence rate (SIR) was 9.1 and the absolute excess risk (AER) was 16.8. Of note, 85% (105 out of 123) patients with SMNs had a tumor in the irradiated region, implying the risk of late sequelae of radiotherapy in HL patients for development of SMN [18].

It is generally accepted that the occurrence of SMN is attributed mostly to genetic factors, environmental factors, and late effects of cancer therapy (particularly chemo- and radiotherapy for the first primaries) [19]. The genetic factors played an important role in SMNs which was supported by the finding of an increased risk of SMNs among lymphoma patients (HL = 7,476; NHL = 25,941) with a family history of cancer, defined as a diagnosis of any cancer in the first‐degree relatives [20]. Compared to HL patients with a negative family history of cancer, a significantly increased risk of breast cancer (RR = 1.81, 95% CI: 1.04–3.16) was found among the HL patients with a positive family history of cancer, while increases of non-melanoma skin cancer (RR = 1.94, 95% CI: 0.86–4.38) and lung cancer (RR = 1.99, 95% CI; 0.73–5.39) were also observed even though the statistical significance was not reached [20]. Heritable diseases and acquiring gene mutations of malignant cells have been suggested as the potential genetic mechanism of SMN. For example, retinoblastoma is frequently caused by inherited mutations of the retinoblastoma gene (RB1), a tumor suppressor gene. In cohort studies of heritable retinoblastomas, the estimated cumulative risk of second bone cancer by 20 years from 3-year survival was 6.0% based on a mean follow-up period of 13.7 years [21] and 7.2% based on a mean follow-up period of 17.6 years [22], respectively. In a retrospective study of retinoblastoma patients, the cumulative risk of second cancer within 20 years of bilateral or heritable retinoblastoma clustered was around 10%, and the predominant types of tumors observed were bone and soft-tissue sarcomas [23]. Other susceptibility genes and associated diseases such as NF1 gene (Recklinghausen's disease), WT1 gene (Wilms' tumour), gene APC (adenomatous polyposis coli), and ATM gene (ataxia telangiectasia) could also contribute to SMNs in the patients with family histories [24]. As only limited studies focus on the genetic factors of SMNs in lymphomas, further study is needed to study the genetic factors of SMN, in particularly to investigate the heritable diseases and associated genes to elucidate the potential mechanism(s) of SMN carcinogenesis in lymphoma conditions.

Environmental and occupational factors are also associated with an increased risk of lymphomas [25, 26]. Studies indicated that there is a high risk of NHL associated to environmental/occupational exposures, with either historically exposed chemicals or specific groups of chemicals such as pesticides and herbicides. Some environmental/occupational exposed chemicals such as insecticides could apply to all the NHL subtypes, while others might be for specific lymphoma subtypes only, e.g., benzene applied to follicular lymphoma [26, 27]. Of note, selection of pesticide chemical types should be considered carefully, e.g., it was found an elevated risk in NHL among the farmers who reported ever being exposed to terbufos, but a decreased risk with organochlorine insecticides and phenoxy herbicides, based on a meta-analysis of farmers from three cohort studies conducted by Consortium of Agricultural Cohort Studies (AGRICOH) [28]. Interestingly, the risk of myeloid malignancies decreased with specific animal species e.g., pig farming where the individuals were exposed to high concentrations of bioaerosols like organic dust in addition to pesticides, however increased risk of myeloproliferative neoplasms was seen with increasing number of sheep/goats [29]. In addition to the farming occupation, the increased risk of NHL subtypes is also associated with the industrial pollutants and productions, including polychlorinated biphenyl [30], trichloroethylene [31], hair dye [32], etc. Several review articles have contributed greatly to the current knowledge on the roles of environmental exposures to the etiology and molecular pathogenesis in lymphomas as well as lymphoma subtypes [33,34,35]. Environmental/occupational determinants have been associated with increased SMNs [19], however the analytic studies in terms of increased risk of SMNs in a cohort of lymphoma patients are not available yet. Further large-scale studies on environmental epidemiology and basic science research to elucidate the molecular mechanisms of chemical exposures on the lymphomagenesis-associated SMNs are urgently.

The late effects of lymphoma therapy in regard to the increased risk of SMNs have been well-studied. Based on individual patient data (IPD) from patients treated for newly diagnosed HL, a meta-analysis was performed to investigate the possible changes in the risk of SMNs [36]. It was found that consolidating radiotherapy was associated with an increased rate of SMNs, while optimizations of treatments such as fewer chemotherapy cycles and reduction of the radiation field/dose did not markedly affect the SMNs [36]. The risk of secondary acute myeloid leukaemia and myelodysplastic syndrome (AML/MDS) was also increased even though efficacy was improved among patients treated with intensified chemotherapy protocols, suggesting the importance to make treatment decisions which could be tailored for individual patients [36]. In a 20-year retrospective follow-up study, total of 1,347 lymphoma patients being treated with a high-dose sequential (HDS) program were analyzed for the cumulative incidence of SMNs [37]. The results showed that the cumulative incidence of secondary myelodysplasia/acute leukemia (sMDS/AL) was 3.09% for 5 years and 4.52% for 10 years. In addition to the secondary hematologic malignancy, the development of secondary solid tumors in lymphoma patients was called attention, especially in the patients treated with monoclonal antibody rituximab which was reported as an independent risk factor for solid tumor development and the cumulative incidence of solid tumors was 2.54% for 5 years and 6.79% for 10 years, respectively [37]. In a study of HL survivors being treated with abdominal radiotherapy or procarbazine-containing chemotherapy, a fivefold increased risk of developing colorectal cancer (CRC) was not only related to therapy but also related to a somatic gene mutation such as the mutations of mismatch repair genes [38]. Haematopoietic stem-cell transplantation (HSCT) was wildly reported for the patients at increased risk of developing SMN [39,40,41,42,43,44,45], i.e., one study reporting an RR of 12.0 (95% CI 8.2–14.5) [46]. The literature about the HSCT-related SMNs has been reviewed in detail recently [47]. In lymphoma survivors, solid cancers including breast cancer, thyroid cancer, lung cancer, gastrointestinal cancers, and sarcomas [48, 49] account for the majority of SMNs, while the most common SMN in HL survivors is secondary breast cancer [50, 51]. Accumulating evidence suggests that the therapy-related risk of SMNs increases after a delay of about 10 to 15 years and depends on age at treatment, patient characteristics, differences in radiotherapy and chemotherapy, radiation dose, and length of follow-up [52]. In HL patients, the age at treatment is the most important risk modifying factor for secondary breast cancer development, the younger the patients are, the higher the risk is [53]. The excess risk in younger patients could be related to the increased sensitivity of the mammary gland to the radiation-effect and it was supported by a study in which women (aged ≤ 35) treated with supra-diaphragmatic radiotherapy for HL at high risk when they received radiotherapy closer to the menarche [54]. Radiotherapy field size is also an important issue, lower risk of secondary breast cancer was found in women who received supra-diaphragmatic field radiotherapy not including the axilla compared to those treated with complete mantle-field radiotherapy [55]. Another important issue needs to pay attention to when assessing the risk of SMNs is the dose distribution variations of therapeutic radiation. While high-doses, intermediate-doses, and low-doses radiotherapy showed inconsistent results, assessing the doses being delivered should consider multiple variable parameters such as patient height, weight, size, volume and shape of the tumor, irradiation modalities, etc. The inconsistent results were also found in the chemotherapy when assessing the risk of SMNs because chemotherapy regimens could not be as a sole modality for lymphoma therapy [50, 51]. Likewise, multiple variable parameters as aforementioned should considered to assess the late effects of chemotherapy in lymphoma patients. The risk of second malignant neoplasm in lymphomas and associated risk factors are summarized in Fig. 1.

Fig. 1
figure 1

Schematic diagram for risk of second malignant neoplasm in lymphomas and associated risk factors

Secondary lymphomas

As improvements in survival of solid tumors also influence the potential risk of development of hematological malignancies, it is of interest to note whether there is a higher risk of lymphoma in the patients previously diagnosed with solid tumors than the general population. Previously, a study was performed to determine the risk of the secondary NHL in a cohort of 5484 consecutive children with a variety of malignancies during a period of 27 years [56]. The probability of secondary NHL in this cohort after the diagnosis of the first malignancy was 0.05% (95% CI, 0.01%, 0.2%) at 5–10 years and 0.16% at 15 years (0.04%, 0.63%). Twenty-four children with secondary NHL had initial lymphohematopoietic neoplasms including Hodgkin's disease (n = 18), acute lymphoblastic leukemia (n = 4), and acute myelogenous leukemia (n = 1), while one child had astrocytoma [56]. In Japan, a retrospective cohort study of 2786 patients who were diagnosed with breast cancer between 1970‐1995 (average follow‐up period, 8.6 years) showed that the subjects aged 20–49 years at the time of diagnosis of breast cancer had a significantly increased risk of NHL (O/E = 6.3, 95% CI = 1.7–16.1)[57]. A population-based cohort of 288,390 colorectal cancer (CRC) patients diagnosed between 1973 and 2012 from the database (Surveillance, Epidemiology, and End Results) was retrospectively reviewed to estimate the relative risk for subsequent primary malignancies [58]. In comparison with the general population, CRC patients showed an significantly increased risk of secondary lymphoma (standardized incidence ratios: 0.92) as well as other secondary solid tumors [58]. A study evaluated the occurrence of renal cell cancer (RCC) and hematologic malignancies in 700 individual patients associated with more than 700 pedigrees of families [59]. The results showed a personal history of both RCC and hematologic malignancies for 26 patients in which 13 NHL patients and 4 HL patients were identified. Interestingly, 74 patients with RCC were noted to have 95 family members with hematologic malignancies in which 42 NHL and 12 HL were identified [59]. In China, a cohort of 836 DLBCL patients between 2013 and 2018 was retrospectively studied to investigate the personal history of solid tumors. Total 34 patients were found to have both DLBCL and solid tumors (including liver cancer, stomach cancer, lung cancer, prostate cancer, breast cancer, CRC, renal cell cancer, cervical cancer, ovarian cancer, thyroid cancer, and pancreatic cancer). Among the 34 patients, 30 patients (88%) were diagnosed with solid tumors before DLBCL diagnosis, while 4 patients (12%) were diagnosed simultaneously for a solid tumor and DLBCL [60]. The cohort studies of secondary lymphomas are summarized in Table 1. A secondary lymphoma in HCC patients was also observed [61, 62], however the retrospective cohort study and systematic analysis in regard to the substantial lymphomas in HCC patients were not available. Whereas the etiologic factors such as hepatitis virus infection are associated with both HCC and lymphomas [63,64,65,66], the additional study, therefore, is needed to clarify the association between HCC and secondary lymphomas to enhance the understanding of the oncogenic potential of lymphomas in patients with primary HCC.

Table 1 Secondary lymphomas

The coexistence of lymphomas and HCC has been mostly reported in clinical patients infected with hepatitis virus [67, 68], while cohort studies and case–control studies have consistently reported that the increased risks of both HCC and lymphoma are associated with infection of HCV or HBV [69,70,71,72]. A Danish HCV cohort (DANVIR) cohort study of liver cancer and non-Hodgkin lymphoma was performed in HCV-infected patients [73]. The results indicated that the 10-year risks for HCC and NHL in HCV-infected patients were 1.0% [95% CI: 0.8–1.3%] and 0.1% (95% CI: 0.1–0.2%), respectively [73]. Compared to general population, the HCV infected patients had a 62.91-fold increased risk of HCC (95% CI: 28.99–136.52), a 29.97-fold increased risk of NHL during the first year of follow-up (95% CI: 6.08–147.84), but a 1.26-fold increased risk of NHL after the first year (95% CI: 0.36–4.41) [73]. A greater propensity has been found for patients with HCV infection to develop NHL, especially MZL and DLBCL, but not FL or T cell lymphomas [74]. An etiopathological role of HCV in lymphoproliferative disorder has been previously described in several studies by the supportive data of the HCV-associated NHL in response to interferon (IFN) and ribavirin therapy [75, 76]. The known major site of HCV replication is in the liver parenchyma, but HCV RNA has been also detected in the peripheral blood mononuclear cells (PBMCs) of infected individuals, including B cells and T cells [77, 78]. As regards HCV associated lymphomagenesis, experimental data support either a direct transformation mechanism or an indirect transformation mechanism. The receptors including CD81, the scavenger receptor SR‐BI, and tight‐junction protein Claudin‐1 have been found to mediate directly HCV entry into cells [79,80,81]. Although an in vitro study was failed to demonstrate that HCV can infect PBMCs, the possibility still remains for HCV‐specific B cells to capture HCV RNA from circulation via anti‐HCV antibodies and rheumatoid factor [82, 83]. HCV entry and active HCV replication in PBMCs was supported by the study in which nearly half of HCV-positive mixed cryoglobulinemia (MC) patients (versus individuals without MC) was detected the HCV minus-strand RNA which required the cellular machinery to synthesize from the positive strand of viral DNA [84]. When lymphocytes infected with HCV, the HCV core protein (C) and non-structural protein 3 (NS3) could induce nitric oxide synthase (NOS) which led to somatic mutations of both oncogenes and tumor suppressor genes contributing to lymphomagenesis [85, 86]. On the other hand, direct HCV infection in lymphocytes might not be necessary because HCV could also serve as an indirect transformation agent to induce somatic mutations of both oncogenes and tumor suppressor genes. For example, an antigenic selection driven maturation process had been suggested for the underlying lymphomagenesis in MC and HCV-associated NHL patients [87, 88]. The HCV envelope glycoprotein E2 would not require direct infection of B cells by HCV because E2 is only expressed on the virion surface. This glycoprotein E2 could bind to B cells via CD81 which is associated with CD19 and CD21 to form a complex, while the CD81-CD19-CD21 complex signaling and B-cell receptor cross-activation were thought to promote B-cell proliferation and NHL development [89, 90].

Contrary to HCV, the HBV infection in lymphoma patients was investigated less intensively even though the association between HBV infection and chronic lymphoproliferative disorders was reported in 1977 [91] much earlier than the reports on HCV in NHL patients. In respect to an etiologic role of HBV in lymphoma, there was a large number of experimental findings indicating that HBV could infect and replicate in the lymphoid cells [92]. Indeed, extrahepatic sites of HBV nucleic acids had been found in lymph nodes, spleen, gonads, thyroid gland, kidneys, pancreas, and adrenal glands from patients with acute HBV infection [93]. Nevertheless, the clinical implication of HBV activity in lymphomagenesis remained a matter of debate. Previously, it was reported that HBV-infected patients showed a 2–threefold higher risk to develop NHL, compared to non-infected patients [94]. However, a cohort study of patients infected with HBV from the Danish Cancer Registry a 17-fold higher risk of HCC for HBV-infected individuals but the risk of NHL was not higher in the HBV-infected cohort compared to non-HBV infected [72]. In a recent study, a cohort of seventy‐two patients with current or resolved HBV infection and B‐NHL between 2000 and 2017 were evaluated for the serological indicators of HBV activity [95]. The results indicated that the serological viral activity of HBV was significantly higher in indolent B‐NHL than aggressive B‐NHL [95]. The secondary lymphomas in primary malignancies and etiologic risk factors are summarized in Fig. 2.

Fig. 2
figure 2

The schematic diagram for risk of secondary lymphomas in primary malignancies and etiologic risk factors of HCV and HBV

Lymphoma and metabolic disorders/diseases

Excess weight and obesity have been linked to many types of cancers including lymphoid neoplasms [96]. Previous meta-analyses show that a greater body mass index (BMI) may increase the risk of HL and NHL [97,98,99,100]. Latterly, additional prospective studies with a large number of cases have been published. In a systematic review and meta-analysis with over 20,000 NHL cases, anthropometric factors are correlated to the NHL risk, in which chronic lymphocytic leukemia/small lymphocytic lymphoma is positively associated with BMI in early adulthood and with height, whereas follicular lymphoma (FL) was only positively associated with height. The increased NHL risk with taller stature may reflect cumulative exposure to hormones/growth factors and nutrition status in early life but many other potential confounders need to be clarified [101]. An updated systematic review of published prospective studies is reported recently. The summary relative risk (RR) per 5 kg/m2 increase in BMI were 1.12 [95% confidence interval (CI): 1.05–1.20] for HL, 1.05 (95% CI: 1.03–1.08) for NHL, 1.11 (95% CI: 1.05–1.16) for DLBCL, respectively [102]. The waist-to-hip ratio was associated with an increased risk of DLBCL by 12% [102]. Although the pathophysiological and biological mechanisms linking obesity and risk of lymphoma are largely unknown, the most studied candidate systems include (i) adipocytokines, (ii) insulin/insulin-like growth factors (IGFs), and (iii) inflammation. It has been found that body fat can lead to changes in circulating levels of adipocytokines such as adiponectin and leptin, which can affect insulin resistance and inflammation [103,104,105]. Obesity may increase the risk of lymphoma by affecting insulin resistance and hyper-insulinemia which leads to increased bio-available insulin-like growth factor-I (IGF-I), a well-known growth factor that promotes cell proliferation and inhibits apoptosis [103].

Diabetes mellitus (DM) is a common metabolic disease and is predicted to be one of the five leading disease-burden contributors by 2030 [106]. There is increasing evidence that insulin and IGF-1 are involved in cellular events such as proliferation and metastasis, suggesting that DM is associated with the development and progression of malignancy [107]. Several previous meta-analyses have been performed previously to investigate the association between DM and NHL risk [108,109,110], and a positive association between DM and risk of NHL is suggested. However, the results from previous studies remain inconsistent because of methodological limitations in terms of the included case–control studies. A recent meta-analysis of 35 cohort studies demonstrated that the association between DM and NHL was much more substantial in an Asian population, while sensitivity analyses suggested the robustness of a positive association between DM and NHL risk [111]. Studies also reported the effect of metformin, one of the most commonly used medications for the treatment of type 2 DM (T2DM), on the outcome in T2DM patients suffering from lymphoma. In a study with a small sample size using the Computerized Patient Record System at a Veterans Affairs Medical Center, significantly greater long-term survival was found in the metformin group (18 patients) than the non-metformin group (20 patients) in lymphoma (5.89 vs 1.29 years, P < 0.001)[112]. In a large cohort study of 610,089 newly diagnosed T2DM patients with 2 or more times of prescription of antidiabetic drugs during 1999–2009, the NHL incidence was followed up until the end of 2011. The results indicated that the use of metformin was associated with a lower risk of NHL compared with non-metformin antidiabetic drugs [113]. Although some reports indicated that metformin might improve outcomes of lymphoma patients, the results from the previous studies showed inconsistent. In a cohort study of newly diagnosed DLBCL (n = 869) and FL (n = 895) patients enrolled in the Mayo component of the Molecular Epidemiology Resource cohort study between 2002 and 2015, the results showed that use of metformin was not associated with improved outcomes in newly diagnosed DLBCL and FL [114]. Metformin has been repeatedly shown to regulate lipid metabolism, not only via its antidiabetic effect but also through the activation of adenosine monophosphate kinase (AMPK) which mediates the regulations of peroxisomal proliferator-activated receptor α (PPARα) and PPARγ [115,116,117]. Studies have shown that PPARα agonists such as fenofibrate and clofibrate significantly reduce cell viability and induce apoptosis in lymphoma cell lines [118, 119]. PPARγ activation contributes to the survival of T lymphoma cells by affecting cellular metabolism [120]. The use of PPARγ ligands could inhibit proliferation and induce apoptosis in MCL [121]. The PPARγ ligands-induced apoptosis was also found in B lymphocytes, Burkitt’s B cell lymphoma cells [122], and DLBCL cells through a PPARγ -independent pathway [123]. A study reported the inhibition of lymphoma cell proliferation by PPARγ ligands via wingless-related integration site (Wnt) signaling pathway [124], which was well accepted as a potential mechanism contributing to lymphomagenesis. Apoptosis induction by PPARγ ligands is also considered for the importance of immune activity, e.g., a study has shown that engagement of CD40 to deliver a potent prosurvival signal prevents the PPARγ agonist-induced apoptosis of B lymphocytes via an NF-kappaB-dependent mechanism [125].

A study, based on multiple pathway crosstalk networks (PCNs), was performed recently to investigate the potential pathways in primary mediastinal B-cell lymphoma (PMBL) via analyzing the data of gene expression, pathway, and protein–protein interaction [126]. In this study, nonalcoholic fatty liver disease (NAFLD) was identified as a pathological mechanism of PMBL and to be one of the most important five hub pathways [including NAFLD, tuberculosis, human T-lymphotropic virus type-I (HTLV-I) infection, hepatitis B, and Epstein-Barr virus infection][126]. Nonalcoholic steatohepatitis (NASH), defined as the liver manifestation of metabolic syndrome and chronic inflammation, is the most severe form of NAFLD [127]. Evidence indicates that NASH patients are at high risk for HCC as well as a variety of other cancers [128]. In fact, liver involvement is not a rare condition in patients with lymphoma including HL, DLBCL, BL, T-cell lymphomas, and marginal zone B-cell lymphomas (MZL)[129]. Therefore, NASH characterized by chronic liver inflammation may represent a possible mechanistic link with lymphomagenesis which deserved further investigation. Although primary hepatic lymphoma (PHL) is a rare clinical entity comprising 0.016% of all cases of NHL and 0.4% of extranodal NHL [130], a substantial number of primary hepatic mucosa-associated lymphoid tissue (MALT) lymphomas were reported to occur in patients with chronic hepatitis or with primary biliary cirrhosis [131,132,133], suggesting a link between chronic liver inflammation and hepatic MALT lymphomagenesis. A case of hepatic MALT lymphomas with NASH diagnosis was reported [134]. On the other hand, the cases were reported in which NASH occurred in the patient with T-lymphoblastic lymphoma during chemotherapy including prednisolone [135] and fatty liver developed in children with non-Hodgkin lymphoma [136]. NASH was induced by induction chemotherapy for pediatric acute lymphoblastic leukemia [137], and even fulminant hepatic failure developed in a woman diagnosed with stage IV Hodgkin’s disease with a short course of prednisone (2 weeks) treatment while post-mortem demonstrated NASH and trivial liver involvement of lymphoma [138]. In a study involving 227 lymphoma patients to follow steatosis using a cut-off value of 42 Hounsfield units on the unenhanced CT part of PET/CT examinations, hepatic steatosis was observed in 11.9% of the patients at some point during their baseline or post-treatment evaluation [139].

The metabolic diseases and associated signaling transductions related to the risk of lymphomas are summarized in Fig. 3.

Fig. 3
figure 3

Schematic diagram of metabolic diseases and associated signaling transductions related to the risk of lymphomas

Obese, diabetes and NASH can lead to metabolic abnormalities of adipose tissue, affecting the release of hormones, adipokines, inflammatory cytokines, growth factors, and free fatty acids [140]. All these metabolic substrates have been implicated as risk factors for lymphomas [141, 142]. Metabolic abnormalities of adipose tissue are associated with chronic inflammation which promotes the production of proinflammatory factors, such as TNF-α, IL-6, and IL-8, and inhibits the secretion of anti-inflammatory factors, such as adiponectin [143, 144]. Adiponectin, an adipose tissue specific cytokine that has a protective role against metabolic disturbances in diabetes and obesity, have been reported to suppress lymphoma growth in mice by modulating NK cells, CD8 T cells, and myeloid-derived suppressor cells [145]. Rimming of adipocytes by lymphoma cells has been recognized in several different entities of lymphoma with skin involvement [146]. The interaction between obese adipocytes and cancer cells can leads to transformation of adipocytes into cancer-associated adipocytes (CAAs) which may promote lymphoma via secreting more leptin and reducing the production of adiponectin [147]. In NASH, chronic liver inflammation causes lipolysis and excessive free fatty acids (FFAs) which are associated with hepatic infiltration of T helper 17 (Th17) cells [148] which mediate the progression from NASH to HCC [149, 150]. Besides its established role of NASH-HCC transition [151], NASH could act as a possible inflammatory trigger of lymphomagenesis, e.g., the breast implant-associated anaplastic large cell lymphoma cell lines and clinical specimens reveal a prominent Th1/Th17 phenotype in advanced disease [152]. In addition, reprogramming of fatty acid metabolism in NASH condition could also contribute to lymphomagenesis. For examples, upregulation of fatty acid synthase (FASN) and de novo lipogenesis are found to associate with the development to a more aggressive phenotype of NHL [153, 154] while inhibition of FASN can induced apoptosis in lymphoma cells and improving antitumor efficacy of chemotherapy [155, 156]. Although evidence suggests a close relationship between metabolic abnormalities and development of lymphomas, it is largely unknown about the lymphomagenesis in obesity, diabetes and NASH. Many important questions remain regarding the microenvironment of metabolic disorder in promoting lymphoma development and progression. Therefore, a more in-depth understanding is required to study, (1) the adipocytes and immune cells in metabolic tumor microenvironment interacting with lymphoma cells and contributing to lymphoma development and progression; (2) identification of specific molecular targets in metabolic pathways to limit tumor proliferation; and (3) determination of new strategies to block the crosstalk between lymphoma cells and tumor associated cells in metabolic tumor microenvironment.

In summary, SMNs remain critical research fields with regard to the etiopathogenesis, carcinogenesis, and optimal management. Therefore, the special emphasis is given to the following important areas. First, it is largely unknown for the genetic factors of SMNs in lymphomas, further study is needed to investigate, in particularly, the heritable diseases and associated genes to elucidate the potential mechanism(s) of SMN carcinogenesis in lymphomans. Second, although large-scale study is important to reveal the potential environmental epidemiology, it is needed to establish the experimental models to elucidate the molecular mechanisms of chemical exposures on the lymphomagenesis-associated SMNs. Third, the late effects of therapy remain the most important issue need to be solved because appropriate clinical manipulation for cancer survivors can prevent or even reverse the carcinogenic process. Multiple variable parameters such as patient height, weight, and size, volume and shape of the tumor should be considered to decide the irradiation/chemo modalities. Last, evidence suggests a close relationship between metabolic abnormalities and development of lymphomas, however it is largely unknown about the lymphomagenesis in the disease conditions such as obesity, diabetes and NASH. More studies are needed to gain insight into the lymphomagenesis linking to chronic inflammation and lipid metabolism which affect inflammatory cytokines, release of hormones, growth factors, adipokines, and free fatty acids. Moreover, as the oncology community applies more cutting-edge technologies such RNA sequencing molecular tools to reveal potential diagnostic and therapeutic targets, a systematic approach to the collection of data on SMNs should be incorporated into the prospective cohort studies of cancer patients.

Availability of data and materials

N/A.

References

  1. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, Advani R, Ghielmini M, Salles GA, Zelenetz AD, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wong JYC, Filippi AR, Dabaja BS, Yahalom J, Specht L. Total body irradiation: guidelines from the International Lymphoma Radiation Oncology Group (ILROG). Int J Radiat Oncol Biol Phys. 2018;101(3):521–9.

    Article  PubMed  Google Scholar 

  3. Carmel RJ, Kaplan HS. Mantle irradiation in Hodgkin’s disease. An analysis of technique, tumor eradication, and complications. Cancer. 1976;37(6):2813–25.

    Article  CAS  PubMed  Google Scholar 

  4. Abdelsalam M, El-Husseiny G, Akhtar S, Khafaga Y, Al-Shabana M, AlHusaini H, El Weshi A, Rahal M, Maghfoor I. Improved survival with combined chemo-radiotherapy in primary central nervous system lymphoma. Hematol Oncol Stem Cell Ther. 2010;3(3):128–34.

    Article  CAS  PubMed  Google Scholar 

  5. Gutterman J, Rodriguez V. Combination chemotherapy of advanced lymphoma. Mil Med. 1972;137(7):255–60.

    Article  CAS  PubMed  Google Scholar 

  6. Rich DC, Corpron CA, Smith MB, Black CT, Lally KP, Andrassy RJ. Second malignant neoplasms in children after treatment of soft tissue sarcoma. J Pediatr Surg. 1997;32(2):369–72.

    Article  CAS  PubMed  Google Scholar 

  7. Ozaki T, Sugihara S, Inoue H. Second malignant neoplasms after treatment for osteosarcoma: a report of three cases. Acta Med Okayama. 1993;47(2):129–33.

    CAS  PubMed  Google Scholar 

  8. Klein G, Michaelis J, Spix C, Wibbing R, Eggers G, Ritter J, Kaatsch P. Second malignant neoplasms after treatment of childhood cancer. Eur J Cancer. 2003;39(6):808–17.

    Article  CAS  PubMed  Google Scholar 

  9. Feig SA. Second malignant neoplasms after successful treatment of childhood cancers. Blood Cells Mol Dis. 2001;27(3):662–6.

    Article  CAS  PubMed  Google Scholar 

  10. de Vathaire F, Hawkins M, Campbell S, Oberlin O, Raquin MA, Schlienger JY, Shamsaldin A, Diallo I, Bell J, Grimaud E, et al. Second malignant neoplasms after a first cancer in childhood: temporal pattern of risk according to type of treatment. Br J Cancer. 1999;79(11–12):1884–93.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Green DM, Hyland A, Barcos MP, Reynolds JA, Lee RJ, Hall BC, Zevon MA. Second malignant neoplasms after treatment for Hodgkin’s disease in childhood or adolescence. J Clin Oncol. 2000;18(7):1492–9.

    Article  CAS  PubMed  Google Scholar 

  12. Attarbaschi A, Carraro E, Ronceray L, Andres M, Barzilai-Birenboim S, Bomken S, Brugieres L, Burkhardt B, Ceppi F, Chiang AKS, et al. Second malignant neoplasms after treatment of non-Hodgkin’s lymphoma-a retrospective multinational study of 189 children and adolescents. Leukemia. 2020;35(2):534–49.

    Article  PubMed  Google Scholar 

  13. Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, Jemal A, Kramer JL, Siegel RL. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 2019;69(5):363–85.

    Article  PubMed  Google Scholar 

  14. Travis LB, Curtis RE, Glimelius B, Holowaty E, Van Leeuwen FE, Lynch CF, Adami J, Gospodarowicz M, Wacholder S, Inskip P, et al. Second cancers among long-term survivors of non-Hodgkin’s lymphoma. J Natl Cancer Inst. 1993;85(23):1932–7.

    Article  CAS  PubMed  Google Scholar 

  15. Tanaka H, Tsukuma H, Teshima H, Ajiki W, Koyama Y, Kinoshita N, Masaoka T, Oshima A. Second primary cancers following non-Hodgkin’s lymphoma in Japan: increased risk of hepatocellular carcinoma. Jpn J Cancer Res. 1997;88(6):537–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mudie NY, Swerdlow AJ, Higgins CD, Smith P, Qiao Z, Hancock BW, Hoskin PJ, Linch DC. Risk of second malignancy after non-Hodgkin’s lymphoma: a British Cohort Study. J Clin Oncol. 2006;24(10):1568–74.

    Article  PubMed  Google Scholar 

  17. Brennan P, Scelo G, Hemminki K, Mellemkjaer L, Tracey E, Andersen A, Brewster DH, Pukkala E, McBride ML, Kliewer EV, et al. Second primary cancers among 109 000 cases of non-Hodgkin’s lymphoma. Br J Cancer. 2005;93(1):159–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dorffel W, Riepenhausenl M, Luders H, Bramswig J, Schellong G. Secondary malignancies following treatment for Hodgkin’s lymphoma in childhood and adolescence. Dtsch Arztebl Int. 2015;112(18):320–7.

    PubMed  PubMed Central  Google Scholar 

  19. Travis LB, Demark Wahnefried W, Allan JM, Wood ME, Ng AK. Aetiology, genetics and prevention of secondary neoplasms in adult cancer survivors. Nat Rev Clin Oncol. 2013;10(5):289–301.

    Article  CAS  PubMed  Google Scholar 

  20. Landgren O, Pfeiffer RM, Stewart L, Gridley G, Mellemkjaer L, Hemminki K, Goldin LR, Travis LB. Risk of second malignant neoplasms among lymphoma patients with a family history of cancer. Int J Cancer. 2007;120(5):1099–102.

    Article  CAS  PubMed  Google Scholar 

  21. Hawkins MM, Draper GJ, Kingston JE. Incidence of second primary tumours among childhood cancer survivors. Br J Cancer. 1987;56(3):339–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hawkins MM, Wilson LM, Burton HS, Potok MH, Winter DL, Marsden HB, Stovall MA. Radiotherapy, alkylating agents, and risk of bone cancer after childhood cancer. J Natl Cancer Inst. 1996;88(5):270–8.

    Article  CAS  PubMed  Google Scholar 

  23. Eng C, Li FP, Abramson DH, Ellsworth RM, Wong FL, Goldman MB, Seddon J, Tarbell N, Boice JD Jr. Mortality from second tumors among long-term survivors of retinoblastoma. J Natl Cancer Inst. 1993;85(14):1121–8.

    Article  CAS  PubMed  Google Scholar 

  24. Kony SJ, de Vathaire F, Chompret A, Shamsaldim A, Grimaud E, Raquin MA, Oberlin O, Brugieres L, Feunteun J, Eschwege F, et al. Radiation and genetic factors in the risk of second malignant neoplasms after a first cancer in childhood. Lancet. 1997;350(9071):91–5.

    Article  CAS  PubMed  Google Scholar 

  25. Piris MA, Medeiros LJ, Chang KC. Hodgkin lymphoma: a review of pathological features and recent advances in pathogenesis. Pathology. 2020;52(1):154–65.

    Article  CAS  PubMed  Google Scholar 

  26. Wong O, Harris F, Armstrong TW, Hua F. A hospital-based case-control study of non-Hodgkin lymphoid neoplasms in Shanghai: analysis of environmental and occupational risk factors by subtypes of the WHO classification. Chem Biol Interact. 2010;184(1–2):129–46.

    Article  CAS  PubMed  Google Scholar 

  27. Morton LM, Holford TR, Leaderer B, Boyle P, Zahm SH, Zhang Y, Flynn S, Tallini G, Zhang B, Owens PH, et al. Cigarette smoking and risk of non-Hodgkin lymphoma subtypes among women. Br J Cancer. 2003;89(11):2087–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Leon ME, Schinasi LH, Lebailly P, Beane Freeman LE, Nordby KC, Ferro G, Monnereau A, Brouwer M, Tual S, Baldi I, et al. Pesticide use and risk of non-Hodgkin lymphoid malignancies in agricultural cohorts from France, Norway and the USA: a pooled analysis from the AGRICOH consortium. Int J Epidemiol. 2019;48(5):1519–35.

    Article  PubMed  PubMed Central  Google Scholar 

  29. El-Zaemey S, Schinasi LH, Ferro G, Tual S, Lebailly P, Baldi I, Nordby KC, Kjaerheim K, Schuz J, Monnereau A, et al. Animal farming and the risk of lymphohaematopoietic cancers: a meta-analysis of three cohort studies within the AGRICOH consortium. Occup Environ Med. 2019;76(11):827–37.

    Article  PubMed  Google Scholar 

  30. Bertrand KA, Spiegelman D, Aster JC, Altshul LM, Korrick SA, Rodig SJ, Zhang SM, Kurth T, Laden F. Plasma organochlorine levels and risk of non-Hodgkin lymphoma in a cohort of men. Epidemiology. 2010;21(2):172–80.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Cocco P, Vermeulen R, Flore V, Nonne T, Campagna M, Purdue M, Blair A, Monnereau A, Orsi L, Clavel J, et al. Occupational exposure to trichloroethylene and risk of non-Hodgkin lymphoma and its major subtypes: a pooled InterLymph [correction of IinterLlymph] analysis. Occup Environ Med. 2013;70(11):795–802.

    Article  CAS  PubMed  Google Scholar 

  32. Linet MS, Vajdic CM, Morton LM, de Roos AJ, Skibola CF, Boffetta P, Cerhan JR, Flowers CR, de Sanjose S, Monnereau A, et al. Medical history, lifestyle, family history, and occupational risk factors for follicular lymphoma: the InterLymph Non-Hodgkin Lymphoma Subtypes Project. J Natl Cancer Inst Monogr. 2014;2014(48):26–40.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Hosnijeh FS, Heederik D, Vermeulen R. A review of the role of lymphoma markers and occupational and environmental exposures. Vet Q. 2012;32(2):61–73.

    Article  PubMed  Google Scholar 

  34. Blinder V, Fisher SG. Lymphoma Research Foundation NY: the role of environmental factors in the etiology of lymphoma. Cancer Invest. 2008;26(3):306–16.

    Article  CAS  PubMed  Google Scholar 

  35. Moubadder L, McCullough LE, Flowers CR, Koff JL. Linking environmental exposures to molecular pathogenesis in non-hodgkin lymphoma subtypes. Cancer Epidemiol Biomarkers Prev. 2020;29(10):1844–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Franklin J, Eichenauer DA, Becker I, Monsef I, Engert A. Optimisation of chemotherapy and radiotherapy for untreated Hodgkin lymphoma patients with respect to second malignant neoplasms, overall and progression-free survival: individual participant data analysis. Cochrane Database Syst Rev. 2017;9:CD008814.

    PubMed  Google Scholar 

  37. Tarella C, Passera R, Magni M, Benedetti F, Rossi A, Gueli A, Patti C, Parvis G, Ciceri F, Gallamini A, et al. Risk factors for the development of secondary malignancy after high-dose chemotherapy and autograft, with or without rituximab: a 20-year retrospective follow-up study in patients with lymphoma. J Clin Oncol. 2011;29(7):814–24.

    Article  CAS  PubMed  Google Scholar 

  38. Rigter LS, Snaebjornsson P, Rosenberg EH, Atmodimedjo PN, Aleman BM, Ten Hoeve J, Geurts-Giele WR, Group P, van Ravesteyn TW, Hoeksel J et al. Double somatic mutations in mismatch repair genes are frequent in colorectal cancer after Hodgkin's lymphoma treatment. Gut 2018; 67(3):447–455.

  39. Rizzo JD, Curtis RE, Socie G, Sobocinski KA, Gilbert E, Landgren O, Travis LB, Travis WD, Flowers ME, Friedman DL, et al. Solid cancers after allogeneic hematopoietic cell transplantation. Blood. 2009;113(5):1175–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ringden O, Brazauskas R, Wang Z, Ahmed I, Atsuta Y, Buchbinder D, Burns LJ, Cahn JY, Duncan C, Hale GA, et al. Second solid cancers after allogeneic hematopoietic cell transplantation using reduced-intensity conditioning. Biol Blood Marrow Transplant. 2014;20(11):1777–84.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Majhail NS, Brazauskas R, Rizzo JD, Sobecks RM, Wang Z, Horowitz MM, Bolwell B, Wingard JR, Socie G. Secondary solid cancers after allogeneic hematopoietic cell transplantation using busulfan-cyclophosphamide conditioning. Blood. 2011;117(1):316–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kasai S, Itonaga H, Niino D, Miyoshi H, Kato T, Imanishi D, Fujioka M, Furumoto T, Sato S, Sawayama Y, et al. Programmed death 1 ligand (PD-L1) in solid cancers after allogeneic hematopoietic stem cell transplantation: a retrospective analysis by the Nagasaki Transplant Group. Int J Hematol. 2020;112(4):524–34.

    Article  CAS  PubMed  Google Scholar 

  43. Gallagher G, Forrest DL. Second solid cancers after allogeneic hematopoietic stem cell transplantation. Cancer. 2007;109(1):84–92.

    Article  PubMed  Google Scholar 

  44. Brown JR, Yeckes H, Friedberg JW, Neuberg D, Kim H, Nadler LM, Freedman AS. Increasing incidence of late second malignancies after conditioning with cyclophosphamide and total-body irradiation and autologous bone marrow transplantation for non-Hodgkin’s lymphoma. J Clin Oncol. 2005;23(10):2208–14.

    Article  CAS  PubMed  Google Scholar 

  45. Witherspoon RP, Fisher LD, Schoch G, Martin P, Sullivan KM, Sanders J, Deeg HJ, Doney K, Thomas D, Storb R, et al. Secondary cancers after bone marrow transplantation for leukemia or aplastic anemia. N Engl J Med. 1989;321(12):784–9.

    Article  CAS  PubMed  Google Scholar 

  46. Bhatia S, Ramsay NK, Steinbuch M, Dusenbery KE, Shapiro RS, Weisdorf DJ, Robison LL, Miller JS, Neglia JP. Malignant neoplasms following bone marrow transplantation. Blood. 1996;87(9):3633–9.

    Article  CAS  PubMed  Google Scholar 

  47. Danylesko I, Shimoni A. Second malignancies after hematopoietic stem cell transplantation. Curr Treat Options Oncol. 2018;19(2):9.

    Article  PubMed  Google Scholar 

  48. Bhatia S, Yasui Y, Robison LL, Birch JM, Bogue MK, Diller L, DeLaat C, Fossati-Bellani F, Morgan E, Oberlin O, et al. High risk of subsequent neoplasms continues with extended follow-up of childhood Hodgkin’s disease: report from the Late Effects Study Group. J Clin Oncol. 2003;21(23):4386–94.

    Article  PubMed  Google Scholar 

  49. van Leeuwen FE, Klokman WJ, Veer MB, Hagenbeek A, Krol AD, Vetter UA, Schaapveld M, van Heerde P, Burgers JM, Somers R, et al. Long-term risk of second malignancy in survivors of Hodgkin’s disease treated during adolescence or young adulthood. J Clin Oncol. 2000;18(3):487–97.

    Article  PubMed  Google Scholar 

  50. Bakkach J, Pellegrino B, Elghazawy H, Novosad O, Agrawal S, Bennani Mechita M. Current overview and special considerations for second breast cancer in Hodgkin lymphoma survivors. Crit Rev Oncol Hematol. 2021;157:103175.

    Article  PubMed  Google Scholar 

  51. Lyman GH, Barron RL, Natoli JL, Miller RM. Systematic review of efficacy of dose-dense versus non-dose-dense chemotherapy in breast cancer, non-Hodgkin lymphoma, and non-small cell lung cancer. Crit Rev Oncol Hematol. 2012;81(3):296–308.

    Article  PubMed  Google Scholar 

  52. Adams MJ, Constine LS, Lipshultz SE. Late effects of therapy for Hodgkin’s lymphoma. Curr Hematol Malig Rep. 2007;2(3):143–50.

    Article  PubMed  Google Scholar 

  53. Ibrahim EM, Abouelkhair KM, Kazkaz GA, Elmasri OA, Al-Foheidi M. Risk of second breast cancer in female Hodgkin’s lymphoma survivors: a meta-analysis. BMC Cancer. 2012;12:197.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Cooke R, Jones ME, Cunningham D, Falk SJ, Gilson D, Hancock BW, Harris SJ, Horwich A, Hoskin PJ, Illidge T, et al. Breast cancer risk following Hodgkin lymphoma radiotherapy in relation to menstrual and reproductive factors. Br J Cancer. 2013;108(11):2399–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schaapveld M, Aleman BM, van Eggermond AM, Janus CP, Krol AD, van der Maazen RW, Roesink J, Raemaekers JM, de Boer JP, Zijlstra JM, et al. Second Cancer Risk Up to 40 Years after treatment for Hodgkin’s Lymphoma. N Engl J Med. 2015;373(26):2499–511.

    Article  CAS  PubMed  Google Scholar 

  56. Eguiguren JM, Ribeiro RC, Pui CH, Hancock ML, Pratt CB, Head DR, Crist WM. Secondary non-Hodgkin’s lymphoma after treatment for childhood cancer. Leukemia. 1991;5(10):908–11.

    CAS  PubMed  Google Scholar 

  57. Tanaka H, Tsukuma H, Koyama H, Kinoshita Y, Kinoshita N, Oshima A. Second primary cancers following breast cancer in the Japanese female population. Jpn J Cancer Res. 2001;92(1):1–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yang J, Li S, Lv M, Wu Y, Chen Z, Shen Y, Wang B, Chen L, Yi M, Yang J. Risk of subsequent primary malignancies among patients with prior colorectal cancer: a population-based cohort study. Onco Targets Ther. 2017;10:1535–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Dutcher JP, Wiernik PH, Varella L, Chintapatla R. Occurrence of renal cell carcinoma and hematologic malignancies (predominantly lymphoid) in individuals and in families. Fam Cancer. 2016;15(4):677–87.

    Article  PubMed  Google Scholar 

  60. Meng YN, Shi Q, Cheng S, Wang L, Zhao WL. Impact of solid tumor history on treatment response and survival of patients with diffuse large B-cell lymphoma. Zhonghua Xue Ye Xue Za Zhi. 2019;40(8):639–43.

    CAS  PubMed  Google Scholar 

  61. Yokohama A, Abe T, Yamada T, Kojima A, Kobayashi I, Ishihara H, Iizuka H, Katagai S, Ito H, Takagi H. A case of jejunal malignant lymphoma resulting in perforation 2 years after transcatheter arterial embolization for hepatocellular carcinoma. Nihon Shokakibyo Gakkai Zasshi. 1996;93(8):578–82.

    CAS  PubMed  Google Scholar 

  62. Ohzato Y, Murakami M, Shimizu J, Koga C, Marukawa D, Yoshida Y, Yasuyama A, Matsumura T, Takada A, Kameda C, et al. A case report of inguinal malignant lymphoma after surgery for hepatocellular carcinoma. Gan To Kagaku Ryoho. 2017;44(12):1638–40.

    PubMed  Google Scholar 

  63. Torres HA, Mahale P. Most patients with HCV-associated lymphoma present with mild liver disease: a call to revise antiviral treatment prioritization. Liver Int. 2015;35(6):1661–4.

    Article  PubMed  Google Scholar 

  64. Hu J, Liu K, Luo J. HIV-HBV and HIV-HCV coinfection and liver cancer development. Cancer Treat Res. 2019;177:231–50.

    Article  CAS  PubMed  Google Scholar 

  65. Fiorino S, Cuppini A, Castellani G, Bacchi-Reggiani ML, Jovine E. HBV- and HCV-related infections and risk of pancreatic cancer. JOP. 2013;14(6):603–9.

    PubMed  Google Scholar 

  66. Borsetto D, Fussey J, Fabris L, Bandolin L, Gaudioso P, Phillips V, Polesel J, Boscolo-Rizzo P. HCV infection and the risk of head and neck cancer: A meta-analysis. Oral Oncol. 2020;109:104869.

    Article  PubMed  Google Scholar 

  67. Lee MH, Lin YC, Cheng HT, Chuang WY, Huang HC, Kao HW. Coexistence of hepatoma with mantle cell lymphoma in a hepatitis B carrier. World J Gastroenterol. 2015;21(45):12981–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lee SI, Heo NY, Park SH, Joo YD, Kim IH, Park JI, Kim JY, Kim SH, Shim HK. Synchronous hepatocellular carcinoma and B-cell non-Hodgkin’s lymphoma in chronic hepatitis C patient. Korean J Gastroenterol. 2014;64(3):168–72.

    Article  PubMed  Google Scholar 

  69. Yeo W, Hui P, Chow JH, Mok TS. Hepatocellular carcinoma and lymphoma–two hepatitis B virus-related malignant diseases. Lancet Oncol. 2001;2(9):543.

    Article  CAS  PubMed  Google Scholar 

  70. Shapira MY, Muszkat M, Braunstein I, Gotsman I. Co-occurrence of hepatocellular carcinoma and lymphoma in patients with hepatitis C virus cirrhosis. J Clin Gastroenterol. 2001;32(4):368–9.

    Article  CAS  PubMed  Google Scholar 

  71. Li K, Wang L, Cheng J, Lu YY, Zhang LX, Mu JS, Hong Y, Liu Y, Duan HJ, Wang G, et al. Interaction between hepatitis C virus core protein and translin protein–a possible molecular mechanism for hepatocellular carcinoma and lymphoma caused by hepatitis C virus. World J Gastroenterol. 2003;9(2):300–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Andersen ES, Omland LH, Jepsen P, Krarup H, Christensen PB, Obel N, Weis N, Study DC. Risk of all-type cancer, hepatocellular carcinoma, non-Hodgkin lymphoma and pancreatic cancer in patients infected with hepatitis B virus. J Viral Hepat 2015; 22(10):828-834

  73. Omland LH, Jepsen P, Krarup H, Christensen PB, Weis N, Nielsen L, Obel N, Sorensen HT, Stuver SO, Study DC. Liver cancer and non-Hodgkin lymphoma in hepatitis C virus-infected patients: results from the DANVIR cohort study. Int J Cancer 2012; 130(10):2310-2317

  74. Persico M, Aglitti A, Caruso R, De Renzo A, Selleri C, Califano C, Abenavoli L, Federico A, Masarone M. Efficacy and safety of new direct antiviral agents in hepatitis C virus-infected patients with diffuse large B-cell non-Hodgkin’s lymphoma. Hepatology. 2018;67(1):48–55.

    Article  CAS  PubMed  Google Scholar 

  75. Hermine O, Lefrere F, Bronowicki JP, Mariette X, Jondeau K, Eclache-Saudreau V, Delmas B, Valensi F, Cacoub P, Brechot C, et al. Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection. N Engl J Med. 2002;347(2):89–94.

    Article  CAS  PubMed  Google Scholar 

  76. Levine AM, Shimodaira S, Lai MM. Treatment of HCV-related mantle-cell lymphoma with ribavirin and pegylated interferon Alfa. N Engl J Med. 2003;349(21):2078–9.

    Article  CAS  PubMed  Google Scholar 

  77. Fournillier A, Freida D, Defrance T, Merle P, Trepo C, Inchauspe G. Analysis of B-lymphocyte differentiation in patients infected with hepatitis C virus. J Med Virol. 2004;72(4):566–74.

    Article  CAS  PubMed  Google Scholar 

  78. Kondo Y, Sung VM, Machida K, Liu M, Lai MM. Hepatitis C virus infects T cells and affects interferon-gamma signaling in T cell lines. Virology. 2007;361(1):161–73.

    Article  CAS  PubMed  Google Scholar 

  79. Meertens L, Bertaux C, Cukierman L, Cormier E, Lavillette D, Cosset FL, Dragic T. The tight junction proteins claudin-1, -6, and -9 are entry cofactors for hepatitis C virus. J Virol. 2008;82(7):3555–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Scarselli E, Ansuini H, Cerino R, Roccasecca RM, Acali S, Filocamo G, Traboni C, Nicosia A, Cortese R, Vitelli A. The human scavenger receptor class B type I is a novel candidate receptor for the hepatitis C virus. EMBO J. 2002;21(19):5017–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, Weiner AJ, Houghton M, Rosa D, Grandi G, et al. Binding of hepatitis C virus to CD81. Science. 1998;282(5390):938–41.

    Article  CAS  PubMed  Google Scholar 

  82. Marukian S, Jones CT, Andrus L, Evans MJ, Ritola KD, Charles ED, Rice CM, Dustin LB. Cell culture-produced hepatitis C virus does not infect peripheral blood mononuclear cells. Hepatology. 2008;48(6):1843–50.

    Article  PubMed  Google Scholar 

  83. Fornasieri A, Bernasconi P, Ribero ML, Sinico RA, Fasola M, Zhou J, Portera G, Tagger A, Gibelli A, D’Amico G. Hepatitis C virus (HCV) in lymphocyte subsets and in B lymphocytes expressing rheumatoid factor cross-reacting idiotype in type II mixed cryoglobulinaemia. Clin Exp Immunol. 2000;122(3):400–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Sansonno D, Tucci FA, Lauletta G, De Re V, Montrone M, Troiani L, Sansonno L, Dammacco F. Hepatitis C virus productive infection in mononuclear cells from patients with cryoglobulinaemia. Clin Exp Immunol. 2007;147(2):241–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Machida K, Cheng KT, Sung VM, Shimodaira S, Lindsay KL, Levine AM, Lai MY, Lai MM. Hepatitis C virus induces a mutator phenotype: enhanced mutations of immunoglobulin and protooncogenes. Proc Natl Acad Sci U S A. 2004;101(12):4262–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Machida K, Cheng KT, Sung VM, Lee KJ, Levine AM, Lai MM. Hepatitis C virus infection activates the immunologic (type II) isoform of nitric oxide synthase and thereby enhances DNA damage and mutations of cellular genes. J Virol. 2004;78(16):8835–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Ivanovski M, Silvestri F, Pozzato G, Anand S, Mazzaro C, Burrone OR, Efremov DG. Somatic hypermutation, clonal diversity, and preferential expression of the VH 51p1/VL kv325 immunoglobulin gene combination in hepatitis C virus-associated immunocytomas. Blood. 1998;91(7):2433–42.

    Article  CAS  PubMed  Google Scholar 

  88. Marasca R, Vaccari P, Luppi M, Zucchini P, Castelli I, Barozzi P, Cuoghi A, Torelli G. Immunoglobulin gene mutations and frequent use of VH1-69 and VH4-34 segments in hepatitis C virus-positive and hepatitis C virus-negative nodal marginal zone B-cell lymphoma. Am J Pathol. 2001;159(1):253–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Machida K, Cheng KT, Pavio N, Sung VM, Lai MM. Hepatitis C virus E2-CD81 interaction induces hypermutation of the immunoglobulin gene in B cells. J Virol. 2005;79(13):8079–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Flint M, McKeating JA. The role of the hepatitis C virus glycoproteins in infection. Rev Med Virol. 2000;10(2):101–17.

    Article  CAS  PubMed  Google Scholar 

  91. Heimann R, Ray MB, Desmet VJ. HBsAg, chronic lymphoproliferative disorders, and cirrhosis of liver. J Clin Pathol. 1977;30(9):817–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Ciesek S, Helfritz FA, Lehmann U, Becker T, Strassburg CP, Neipp M, Ciner A, Fytili P, Tillmann HL, Manns MP, et al. Persistence of occult hepatitis B after removal of the hepatitis B virus-infected liver. J Infect Dis. 2008;197(3):355–60.

    Article  CAS  PubMed  Google Scholar 

  93. Yoffe B, Burns DK, Bhatt HS, Combes B. Extrahepatic hepatitis B virus DNA sequences in patients with acute hepatitis B infection. Hepatology. 1990;12(2):187–92.

    Article  CAS  PubMed  Google Scholar 

  94. Marcucci F, Spada E, Mele A, Caserta CA, Pulsoni A. The association of hepatitis B virus infection with B-cell non-Hodgkin lymphoma—a review. Am J Blood Res. 2012;2(1):18–28.

    PubMed  PubMed Central  Google Scholar 

  95. Zhou X, Wuchter P, Egerer G, Kriegsmann M, Kommoss FKF, Witzens-Harig M, Kriegsmann K. Serological hepatitis B virus (HBV) activity in patients with HBV infection and B-cell non-Hodgkin’s lymphoma. Eur J Haematol. 2020;104(5):469–75.

    Article  CAS  PubMed  Google Scholar 

  96. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and cancer risk: emerging biological mechanisms and perspectives. Metabolism. 2019;92:121–35.

    Article  CAS  PubMed  Google Scholar 

  97. Pan SY, Mao Y, Ugnat AM, Canadian Cancer Registries Epidemiology Research G. Physical activity, obesity, energy intake, and the risk of non-Hodgkin’s lymphoma: a population-based case-control study. Am J Epidemiol. 2005;162(12):1162–73.

    Article  PubMed  Google Scholar 

  98. Larsson SC, Wolk A. Obesity and risk of non-Hodgkin’s lymphoma: a meta-analysis. Int J Cancer. 2007;121(7):1564–70.

    Article  CAS  PubMed  Google Scholar 

  99. Maskarinec G, Erber E, Gill J, Cozen W, Kolonel LN. Overweight and obesity at different times in life as risk factors for non-Hodgkin’s lymphoma: the multiethnic cohort. Cancer Epidemiol Biomarkers Prev. 2008;17(1):196–203.

    Article  PubMed  Google Scholar 

  100. Larsson SC, Wolk A. Body mass index and risk of non-Hodgkin’s and Hodgkin’s lymphoma: a meta-analysis of prospective studies. Eur J Cancer. 2011;47(16):2422–30.

    Article  PubMed  Google Scholar 

  101. Hidayat K, Li HJ, Shi BM. Anthropometric factors and non-Hodgkin’s lymphoma risk: systematic review and meta-analysis of prospective studies. Crit Rev Oncol Hematol. 2018;129:113–23.

    Article  PubMed  Google Scholar 

  102. Abar L, Sobiecki JG, Cariolou M, Nanu N, Vieira AR, Stevens C, Aune D, Greenwood DC, Chan DSM, Norat T. Body size and obesity during adulthood, and risk of lympho-haematopoietic cancers: an update of the WCRF-AICR systematic review of published prospective studies. Ann Oncol. 2019;30(4):528–41.

    Article  CAS  PubMed  Google Scholar 

  103. Dalamaga M, Christodoulatos GS. Adiponectin as a biomarker linking obesity and adiposopathy to hematologic malignancies. Horm Mol Biol Clin Investig. 2015;23(1):5–20.

    CAS  PubMed  Google Scholar 

  104. Askmyr M, Quach J, Purton LE. Effects of the bone marrow microenvironment on hematopoietic malignancy. Bone. 2011;48(1):115–20.

    Article  PubMed  Google Scholar 

  105. Park J, Euhus DM, Scherer PE. Paracrine and endocrine effects of adipose tissue on cancer development and progression. Endocr Rev. 2011;32(4):550–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Rathmann W, Giani G. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27(10):2568–9.

    Article  PubMed  Google Scholar 

  107. Gallagher EJ, LeRoith D. Diabetes, cancer, and metformin: connections of metabolism and cell proliferation. Ann N Y Acad Sci. 2011;1243:54–68.

    Article  CAS  PubMed  Google Scholar 

  108. Mitri J, Castillo J, Pittas AG. Diabetes and risk of Non-Hodgkin’s lymphoma: a meta-analysis of observational studies. Diabetes Care. 2008;31(12):2391–7.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Chao C, Page JH. Type 2 diabetes mellitus and risk of non-Hodgkin lymphoma: a systematic review and meta-analysis. Am J Epidemiol. 2008;168(5):471–80.

    Article  PubMed  Google Scholar 

  110. Castillo JJ, Mull N, Reagan JL, Nemr S, Mitri J. Increased incidence of non-Hodgkin lymphoma, leukemia, and myeloma in patients with diabetes mellitus type 2: a meta-analysis of observational studies. Blood. 2012;119(21):4845–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Wang Y, Liu X, Yan P, Bi Y, Liu Y, Zhang ZJ. Association between type 1 and type 2 diabetes and risk of non-Hodgkin’s lymphoma: a meta-analysis of cohort studies. Diabetes Metab. 2019;46(1):8–19.

    Article  PubMed  Google Scholar 

  112. Wynn A, Vacheron A, Zuber J, Solomon SS. Metformin associated with increased survival in type 2 diabetes patients with pancreatic cancer and lymphoma. Am J Med Sci. 2019;358:200–3.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Tseng CH. Metformin is associated with a lower risk of non-Hodgkin lymphoma in patients with type 2 diabetes. Diabetes Metab. 2019;45(5):458–64.

    Article  CAS  PubMed  Google Scholar 

  114. Wang Y, Maurer MJ, Larson MC, Allmer C, Feldman AL, Bennani NN, Thompson CA, Porrata LF, Habermann TM, Witzig TE, et al. Impact of metformin use on the outcomes of newly diagnosed diffuse large B-cell lymphoma and follicular lymphoma. Br J Haematol. 2019;180:820–8.

    Article  Google Scholar 

  115. Salminen A, Kauppinen A, Kaarniranta K. FGF21 activates AMPK signaling: impact on metabolic regulation and the aging process. J Mol Med (Berl). 2017;95(2):123–31.

    Article  CAS  Google Scholar 

  116. Ikhlas S, Ahmad M. Metformin: insights into its anticancer potential with special reference to AMPK dependent and independent pathways. Life Sci. 2017;185:53–62.

    Article  CAS  PubMed  Google Scholar 

  117. Morrison A, Li J. PPAR-gamma and AMPK–advantageous targets for myocardial ischemia/reperfusion therapy. Biochem Pharmacol. 2011;82(3):195–200.

    Article  CAS  PubMed  Google Scholar 

  118. Schmeel LC, Schmeel FC, Schmidt-Wolf IG. Clofibrate demonstrates efficacy in in vitro treatment of lymphoma and multiple myeloma. Anticancer Res. 2016;36(7):3395–400.

    CAS  PubMed  Google Scholar 

  119. Schmeel LC, Schmeel FC, Schmidt-Wolf IGH. In vitro apoptosis induction by fenofibrate in lymphoma and multiple myeloma. Anticancer Res. 2017;37(7):3513–20.

    CAS  PubMed  Google Scholar 

  120. Yang C, Jo SH, Csernus B, Hyjek E, Liu Y, Chadburn A, Wang YL. Activation of peroxisome proliferator-activated receptor gamma contributes to the survival of T lymphoma cells by affecting cellular metabolism. Am J Pathol. 2007;170(2):722–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Eucker J, Sterz J, Krebbel H, Zavrski I, Kaiser M, Zang C, Heider U, Jakob C, Elstner E, Sezer O. Peroxisome proliferator-activated receptor-gamma ligands inhibit proliferation and induce apoptosis in mantle cell lymphoma. Anticancer Drugs. 2006;17(7):763–9.

    Article  CAS  PubMed  Google Scholar 

  122. Ray DM, Akbiyik F, Phipps RP. The peroxisome proliferator-activated receptor gamma (PPARgamma) ligands 15-deoxy-Delta 12,14-prostaglandin J2 and ciglitazone induce human B lymphocyte and B cell lymphoma apoptosis by PPARgamma-independent mechanisms. J Immunol. 2006;177(8):5068–76.

    Article  CAS  PubMed  Google Scholar 

  123. Ray DM, Morse KM, Hilchey SP, Garcia TM, Felgar RE, Maggirwar SB, Phipps RP, Bernstein SH. The novel triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) induces apoptosis of human diffuse large B-cell lymphoma cells through a peroxisome proliferator-activated receptor gamma-independent pathway. Exp Hematol. 2006;34(9):1202–11.

    Article  CAS  PubMed  Google Scholar 

  124. Liu JJ, Dai XJ, Xu Y, Liu PQ, Zhang Y, Liu XD, Fang ZG, Lin DJ, Xiao RZ, Huang RW, et al. Inhibition of lymphoma cell proliferation by peroxisomal proliferator-activated receptor-gamma ligands via Wnt signaling pathway. Cell Biochem Biophys. 2012;62(1):19–27.

    Article  CAS  PubMed  Google Scholar 

  125. Ray DM, Akbiyik F, Bernstein SH, Phipps RP. CD40 engagement prevents peroxisome proliferator-activated receptor gamma agonist-induced apoptosis of B lymphocytes and B lymphoma cells by an NF-kappaB-dependent mechanism. J Immunol. 2005;174(7):4060–9.

    Article  CAS  PubMed  Google Scholar 

  126. Zheng ML, Zhou NK, Luo CH. Utilizing multiple pathway cross-talk networks reveals hub pathways in primary mediastinal B-cell lymphoma. J Cancer Res Ther. 2018;14(Supplement):S694–700.

    Article  CAS  PubMed  Google Scholar 

  127. Michelotti GA, Machado MV, Diehl AM. NAFLD, NASH and liver cancer. Nat Rev Gastroenterol Hepatol. 2013;10(11):656–65.

    Article  CAS  PubMed  Google Scholar 

  128. Bugianesi E. Non-alcoholic steatohepatitis and cancer. Clin Liver Dis. 2007;11(1):191–207.

    Article  CAS  PubMed  Google Scholar 

  129. Baumhoer D, Tzankov A, Dirnhofer S, Tornillo L, Terracciano LM. Patterns of liver infiltration in lymphoproliferative disease. Histopathology. 2008;53(1):81–90.

    Article  CAS  PubMed  Google Scholar 

  130. Masood A, Kairouz S, Hudhud KH, Hegazi AZ, Banu A, Gupta NC. Primary non-Hodgkin lymphoma of liver. Curr Oncol. 2009;16(4):74–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Gherlan GS, Stoia R, Enyedi M, Dobrea C, Calistru PI. Primary hepatic marginal zone lymphoma in a patient with chronic hepatitis C. Maedica (Buchar). 2016;11(3):250–4.

    Google Scholar 

  132. Bao C, Wei J, Zhao X, Lin L, Chen D, Liu K, Qian W, Anas JM, Zhao K. Prognostic value of fluorine-18-fluorodeoxyglucose positron emission tomography/computed tomography in primary hepatic mucosa-associated lymphoid tissue lymphoma: a case report and review of the literature. Medicine (Baltimore). 2018;97(10):e9877.

    Article  Google Scholar 

  133. Dong S, Chen L, Chen Y, Chen X. Primary hepatic extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue type: a case report and literature review. Medicine (Baltimore). 2017;96(13):e6305.

    Article  Google Scholar 

  134. Haefliger S, Milowich D, Sciarra A, Trimeche M, Bouilly J, Kaiser J, Volpi S, Brouland JP, Sempoux C, de Leval L. Primary hepatic marginal B cell lymphoma of mucosa-associated lymphoid tissue (MALT) and non-alcoholic steatohepatitis (NASH): more than a coincidence? Ann Hematol. 2019;98(6):1513–6.

    Article  PubMed  Google Scholar 

  135. Shigematsu A, Okada K, Abe N, Ota S, Kato N, Kondo K, Hige S, Tanaka J, Asaka M, Imamura M. Non-alcoholic steatohepatitis occurring in a patient with T-lymphoblastic lymphoma during chemotherapy including prednisolone. Leuk Lymphoma. 2006;47(7):1397–9.

    Article  PubMed  Google Scholar 

  136. Kose D, Erol C, Kaya F, Koplay M, Koksal Y. Development of fatty liver in children with non-Hodgkin lymphoma. Turk J Pediatr. 2014;56(4):399–403.

    PubMed  Google Scholar 

  137. Yamamoto M, Hori T, Igarashi K, Iesato K, Saito M, Miyanishi K, Kikuchi N, Fujita H, Tsutsumi H. Non-alcoholic steatohepatitis induced by induction chemotherapy for pediatric acute lymphoblastic leukemia. Int J Hematol. 2018;107(4):390–1.

    Article  PubMed  Google Scholar 

  138. Kosmidou IS, Aggarwal A, Ross JJ, Worthington MG. Hodgkin’ s disease with fulminant non-alcoholic steatohepatitis. Dig Liver Dis. 2004;36(10):691–3.

    Article  CAS  PubMed  Google Scholar 

  139. Peters AM, Keramida G, Pencharz D. Assessment of alteration in liver (18)F-FDG uptake due to steatosis in lymphoma patients and its impact on the Deauville score. Eur J Nucl Med Mol Imaging. 2018;45(12):2231–2.

    Article  CAS  PubMed  Google Scholar 

  140. Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 2019;129(10):3990–4000.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Oka T, Mizuno H, Sakata M, Fujita H, Yoshino T, Yamano Y, Utsumi K, Masujima T, Utsunomiya A. Metabolic abnormalities in adult T-cell leukemia/lymphoma and induction of specific leukemic cell death using photodynamic therapy. Sci Rep. 2018;8(1):14979.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Cabanillas F. Metabolic abnormalities in lymphoma. Clin Lymphoma. 2002;3(Suppl 1):S32-36.

    Article  PubMed  Google Scholar 

  143. Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, Beguinot F. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front Physiol. 2019;10:1607.

    Article  PubMed  Google Scholar 

  144. Wang X, Rao H, Liu F, Wei L, Li H, Wu C. Recent advances in adipose tissue dysfunction and its role in the pathogenesis of non-alcoholic fatty liver disease. Cells. 2021;10(12):3300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Han S, Jeong AL, Lee S, Park JS, Kim KD, Choi I, Yoon SR, Lee MS, Lim JS, Han SH, et al. Adiponectin deficiency suppresses lymphoma growth in mice by modulating NK cells, CD8 T cells, and myeloid-derived suppressor cells. J Immunol. 2013;190(9):4877–86.

    Article  CAS  PubMed  Google Scholar 

  146. Lozzi GP, Massone C, Citarella L, Kerl H, Cerroni L. Rimming of adipocytes by neoplastic lymphocytes: a histopathologic feature not restricted to subcutaneous T-cell lymphoma. Am J Dermatopathol. 2006;28(1):9–12.

    Article  PubMed  Google Scholar 

  147. Zhao C, Wu M, Zeng N, Xiong M, Hu W, Lv W, Yi Y, Zhang Q, Wu Y. Cancer-associated adipocytes: emerging supporters in breast cancer. J Exp Clin Cancer Res. 2020;39(1):156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Rau M, Schilling AK, Meertens J, Hering I, Weiss J, Jurowich C, Kudlich T, Hermanns HM, Bantel H, Beyersdorf N, et al. Progression from nonalcoholic fatty liver to nonalcoholic steatohepatitis is marked by a higher frequency of Th17 cells in the liver and an increased Th17/resting regulatory T cell ratio in peripheral blood and in the liver. J Immunol. 2016;196(1):97–105.

    Article  CAS  PubMed  Google Scholar 

  149. Gomes AL, Teijeiro A, Buren S, Tummala KS, Yilmaz M, Waisman A, Theurillat JP, Perna C, Djouder N. Metabolic inflammation-associated IL-17A causes non-alcoholic steatohepatitis and hepatocellular carcinoma. Cancer Cell. 2016;30(1):161–75.

    Article  CAS  PubMed  Google Scholar 

  150. Giles DA, Moreno-Fernandez ME, Stankiewicz TE, Graspeuntner S, Cappelletti M, Wu D, Mukherjee R, Chan CC, Lawson MJ, Klarquist J, et al. Thermoneutral housing exacerbates nonalcoholic fatty liver disease in mice and allows for sex-independent disease modeling. Nat Med. 2017;23(7):829–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Cholankeril G, Patel R, Khurana S, Satapathy SK. Hepatocellular carcinoma in non-alcoholic steatohepatitis: Current knowledge and implications for management. World J Hepatol. 2017;9(11):533–43.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Deva AK, Turner SD, Kadin ME, Magnusson MR, Prince HM, Miranda RN, Inghirami GG, Adams WP Jr. Etiology of breast implant-associated anaplastic large cell lymphoma (BIA-ALCL): current directions in research. Cancers (Basel). 2020;12(12):3861.

    Article  CAS  Google Scholar 

  153. Ravi D, Beheshti A, Abermil N, Lansigan F, Kinlaw W, Matthan NR, Mokhtar M, Passero FC Jr, Puliti P, David KA, et al. Oncogenic integration of nucleotide metabolism via fatty acid synthase in non-hodgkin lymphoma. Front Oncol. 2021;11:725137.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Zhong X, Liu Z, Luo Q, Li J, Zhang W, Shuang Y. Upregulation of fatty acid synthase in MYC and BCL-2 double-expressor lymphoma. Oncol Lett. 2021;21(4):245.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Kant S, Kumar A, Singh SM. Tumor growth retardation and chemosensitizing action of fatty acid synthase inhibitor orlistat on T cell lymphoma: implication of reconstituted tumor microenvironment and multidrug resistance phenotype. Biochim Biophys Acta. 2014;1840(1):294–302.

    Article  CAS  PubMed  Google Scholar 

  156. Kant S, Kumar A, Singh SM. Fatty acid synthase inhibitor orlistat induces apoptosis in T cell lymphoma: role of cell survival regulatory molecules. Biochim Biophys Acta. 2012;1820(11):1764–73.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

N/A.

Funding

This work was supported by Jilin Province Development and Reform Commission (2018SCZWSZX-031).

Author information

Authors and Affiliations

Authors

Contributions

YY, XS, and XW worked literature research and reviewing. YY worked for the analysis of published data from literature and manuscript draft writing. PZ and OB contributed literature research design, clinical consultant and manuscript draft writing. YL contributed to literature research design and manuscript English checking and editing.

Corresponding authors

Correspondence to Ping Zhang, Ou Bai or Yan Li.

Ethics declarations

Ethics approval and consent to participate

N/A.

Consent for publication

All authors have reviewed the manuscript and agreed with for publication.

Competing interests

All authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, Y., Shi, X., Wang, X. et al. Second malignant neoplasms in lymphomas, secondary lymphomas and lymphomas in metabolic disorders/diseases. Cell Biosci 12, 30 (2022). https://doi.org/10.1186/s13578-022-00763-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13578-022-00763-0

Keywords