Next Article in Journal
Is High Expression of Claudin-7 in Advanced Colorectal Carcinoma Associated with a Poor Survival Rate? A Comparative Statistical and Artificial Intelligence Study
Next Article in Special Issue
Pediatric Precursor B-Cell Lymphoblastic Malignancies: From Extramedullary to Medullary Involvement
Previous Article in Journal
Safety and Efficacy of Bevacizumab in Cancer Patients with Inflammatory Bowel Disease
Previous Article in Special Issue
Consensus Recommendations for the Clinical Management of Hematological Malignancies in Patients with DNA Double Stranded Break Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Management of Aggressive Non-Hodgkin Lymphomas in the Pediatric, Adolescent, and Young Adult Population: An Adult vs. Pediatric Perspective

1
Department of Pediatrics, Pediatric Hematology Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
2
Department of Pediatrics, McGovern Medical School, The University of Texas at Houston Health Science Center, Houston, TX 77030, USA
3
Department of Pediatrics, CARTOX Program, Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
4
Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(12), 2912; https://doi.org/10.3390/cancers14122912
Submission received: 13 May 2022 / Revised: 4 June 2022 / Accepted: 8 June 2022 / Published: 13 June 2022
(This article belongs to the Special Issue Non-Hodgkin Lymphoma in Children)

Abstract

:

Simple Summary

This review details the diagnosis and treatment of primary non-Hodgkin lymphoma (NHL) in the pediatric and adolescent population. We also describe treatment modalities such as hematopoietic stem cell transplantation for relapsed or refractory NHL in patients that fail or do not respond to the initial therapy. We then detail the current advancements in treatment for patients that fail initial therapy such as CAR T-cell therapy, the use of immunotherapy that target surface makers on malignant cells and highlight areas where further research is needed. The purpose of our review is to inform the pediatric oncology community in regard to the various types of NHLs and emphasize areas where the science is evolving in the treatment of primary, relapsed or refractory disease.

Abstract

Non-Hodgkin lymphoma (NHL) is a broad entity which comprises a number of different types of lymphomatous malignancies. In the pediatric and adolescent population, the type and prognosis of NHL varies by age and gender. In comparison to adults, pediatric and adolescent patients generally have better outcomes following treatment for primary NHL. However, relapsed/refractory (R/R) disease is associated with poorer outcomes in many types of NHL such as diffuse large B cell lymphoma and Burkitt lymphoma. Newer therapies have been approved in the use of primary NHL in the pediatric and adolescent population such as Rituximab and other therapies such as chimeric antigen receptor T-cell (CAR T-cell) therapy are under investigation for the treatment of R/R NHL. In this review, we feature the characteristics, diagnosis, and treatments of the most common NHLs in the pediatric and adolescent population and also highlight the differences that exist between pediatric and adult disease. We then detail the areas of treatment advances such as immunotherapy with CAR T-cells, brentuximab vedotin, and blinatumomab as well as cell cycle inhibitors and describe areas where further research is needed. The aim of this review is to juxtapose established research regarding pediatric and adolescent NHL with recent advancements as well as highlight treatment gaps where more investigation is needed.

1. Introduction

Non-Hodgkin lymphoma (NHL) encompasses a complex set of lymphomatous tumors. More than 50 different types of NHL have been classified by the World Health Organization [1,2]. The prevalence, outcomes, and treatments of NHL differ depending on its type [3]. Moreover, it is important to distinguish between pediatric and adolescent NHLs and those seen in adults because diagnostic markers and outcomes can differ widely between the two populations. While children with most common types of NHL have excellent event-free survival (EFS) and overall survival (OS) rates, those with relapsed or refractory (R/R) NHL have poor outcomes despite advances in salvage chemotherapy and hematopoietic stem cell transplantation (HSCT). Moreover, current treatment regimens for NHL are associated with adverse effects such as mucositis, decreased appetite, and risks to fertility [4]. Recent advances in the treatment of NHL, such as the combination of rituximab, an anti-CD20 monoclonal antibody, with multiagent chemotherapy regimens or the use of brentuximab, an anti-CD30 antibody-drug conjugate, have improved EFS and OS rates in pediatric and adolescent patients with primary NHL [5]. In this literature review, we discuss the molecular and clinical characteristics of pediatric and adolescent NHL and compare them to those seen in adults with NHL. We then discuss the conventional treatment of primary NHL as well as that of R/R disease. We end the review by describing recent advances in the treatment of NHL, including immunotherapies such as chimeric antigen receptor (CAR) T-cell therapy and cell cycle checkpoint inhibition, and highlighting areas that require further research, for instance whether targeted drugs can improve survival rates of patients with R/R disease.

2. Epidemiology of Non-Hodgkin Lymphoma in Pediatric and Adolescent Patients

The most common types of NHL observed in the pediatric population include Burkitt lymphoma, diffuse large B-cell lymphoma (DLBCL), lymphoblastic lymphoma (both B and T cell origin) while less common types such as anaplastic large-cell lymphoma (ALCL), primary mediastinal large B-cell lymphoma (PMLBCL), and follicular lymphoma are also seen [6]. The type and prevalence of NHL in children and adolescent varies by age, and outcomes depend on both age and sex [7]. Two of the most common types of NHL in the pediatric population are Burkitt lymphoma, which arise from B lymphocytes, and lymphoblastic lymphoma, of which nearly 80% arise from T cells, with the rest originating from B cells, especially in patients from 1 to 9 years old. As children age, the incidence of Burkitt lymphoma decreases, whereas the incidence of DLBCL markedly increases, especially in 15–19-year-olds [6]. In those less than 20 years of age, NHL is relatively uncommon, comprising only 7% of all cancers. In children younger than 5, the incidence is about 6 per million and in adolescents about 15 per million [8].
Moreover, outcomes are markedly better in pediatric and adolescent patients with NHL than in adults [6]. Data from the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program show that over a 24-year period, the prevalence of NHL amongst all cancers has increased by more than 30%; however, over a 10-year period, the survival rate of NHL has improved by nearly 20% with all subgroups including infants, pediatrics, and adolescents having a 10-year survival rate of more than 60%. Patients between the ages of 1 and 4 years account for the best improvement, with a 10-year survival rate of 78% [9]. A review of data from 13 SEER registries that included patients younger than 30 found that males were diagnosed with NHL more often than females, with prevalence rates of 60% and 30% of NHL cases, respectively [10]. However, a study of pediatric patients with NHL found that adolescents (15–18 years old) and girls were typically diagnosed with more advanced NHL (>stage II) and that girls had lower 5-year EFS rates than did boys (70% vs. 83%, respectively) [11].

3. Pathophysiology, Clinical Outcomes, and Clinical Characteristics of Pediatric and Adolescent Non-Hodgkin Lymphoma

3.1. Burkitt Lymphoma

Burkitt lymphoma involves intermediate-sized B cells expressing the surface markers CD19, CD20, and immunoglobulin kappa or gamma light chains. These cells can infiltrate nodal or extra-nodal tissue, and their rate of cell division is considered to be the highest of any human tumor. Thus, Burkitt lymphoma is a highly aggressive type of NHL [12,13]. The chromosomal translocation characteristic of Burkitt lymphoma involves the distal region of the long arm of chromosome 14 and the long arm of chromosome 8 (t(8;14)), which leads to translocation of the oncogene MYC to one of the loci encoding for the immunoglobulin heavy chain [14]. This leads to activation of MYC and the malignant transformation of B-type lymphoma cells [15]. Burkitt lymphoma is classified into three types: endemic, sporadic, and immunodeficiency-related (e.g., those associated with HIV infection) [16,17]. Burkitt lymphoma was one of the first malignancies to be associated with a virus when Epstein et al. [18,19] isolated viral particles, later named the Epstein Barr virus (EBV), from lymphomatous cells. The endemic type of Burkitt lymphoma is closely associated with EBV and malaria infections and is especially prevalent in equatorial regions of Africa, South America, and Oceania, such as Papua New Guinea [20,21,22]. The endemic form most commonly affects the jaw, whereas the sporadic form is most often seen in the abdomen followed by the head and neck region (not including the jaw), the bone marrow, and the central nervous system (CNS) [17,23]. The general presentation of patients with BL includes a rapidly enlarging mass, lab values indicating spontaneous tumor lysis, such as increased uric acid or lactate dehydrogenase, and in those with abdominal disease, abdominal distention and nausea/vomiting [17]. Moreover, in pediatric patients outside the common age in which intussusception may be seen, intussusception may be the presenting symptom, requiring surgical excision and the disease can be associated with early-stage disease with favorable response following surgical resection and chemotherapy [24,25].

3.2. Diffuse Large B-Cell Lymphoma

Although DLBCL is the most common NHL in adults, it accounts for about 10–20% of NHLs in the pediatric and adolescent population. The clinical presentation is most consistent with diffuse lymphadenopathy in areas like the head and neck and inguinal regions, hepato-splenomegaly, and B-symptoms including fever, weight loss, and night sweats [26]. Extra-nodal involvement may also include the abdomen followed by the skin, with infiltration of other organs also a possibility. Clinical emergencies may include superior vena cava (SVC) syndrome and compression of the large airways, leading to respiratory distress [26].
Like Burkitt lymphoma, pediatric DLBCL is an aggressive malignancy [23]. However, the EFS rate of pediatric and adolescent patients with DLBCL is nearly 90%, compared in comparison to about 50% in adults, including those treated with rituximab [27,28,29]. In adult DLBCL, germinal center DLBCL is the most common subtype, comprising about 40–50% of cases, and carries a better prognosis than the activated B-cell-like subtype (5-year OS rates of 59% vs. 30%, respectively). In contrast, in the pediatric and adolescent population (from 0 to 19 years of age), more than 70% of patients with NHL have germinal center-type DLBCL. This subtype of DLBCL is characterized by CD10 and BCL6 expression. Moreover, unlike the adult version of DLBCL where the overexpression of BCL2 protein due to t(14;18) occurs in 20–30% of cases, the incidence of the translocation is believed to be much lower in pediatric patients [30,31,32,33].

3.3. Primary Mediastinal Large B-Cell Lymphoma

Primary MLBCL (PMLBCL) was once considered a type of DLBCL but is now considered a distinct entity in the NHL class and makes up nearly 2% of NHLs in patients younger than 18 [1,34]. The clinical presentation is most likely to manifest with involvement of the upper mediastinum, leading to involvement of the chest wall, heart, lungs, and pleura with associated pleura/pericardial effusions [35]. Similar to DLBCL, PMLBCL may also be associated with SVC syndrome and respiratory distress due to airway compression [36].
PMLBCL been found to be associated with gain-of-function mutations involving chromosome 9, where the genes JAK2, PDL1, and PDL2 are located, as well as mutations affecting the STAT pathway that lead to constitutive STAT6 activation. Activation of STAT6 can lead to evasion of apoptosis and promotion of cell proliferation [23,37,38]. The pathologic characteristics of PMLBCL tumors can vary; some cells show pale cytoplasm, moderately sized nuclei, and sclerosis that overlaps with features of classical Hodgkin lymphoma. Cell surface markers include CD19, CD20, CD30, and CD79a [37,39]. While primary involvement of the mediastinum, including the lungs, pleura, and pericardium, is common, and spread to organs such as the kidneys or liver may occur, CNS and bone marrow involvement is generally rare [40,41]. However, advancements in genetic testing through methods such as fluorescence in situ hybridization have facilitated detection of CIITA and PDL1/PDL2 genes in cases of extra-mediastinal PMLBCL that were otherwise classified as DLBCL, indicating that tumor genetic testing may help recognize other cases of extra-mediastinal PMLBCL erroneously classified as other NHLs [42]. PMLBCL generally carries a favorable prognosis, as shown by a Children’s Cancer Group (CCG) study in which 100% of patients demonstrated a complete response (CR), despite exposure to three different regimens, with EFS and OS rates of 75% and 85%, respectively. However, these rates are worse than those in localized large-cell lymphoma, which had EFS and OS rates of 92% and 97%, respectively [43].

3.4. Lymphoblastic Lymphoma

Lymphoblastic lymphoma is the second most common NHL in children, accounting for nearly a quarter to a third of all pediatric NHL cases with a bimodal age of distribution, seen at a median age of 1–4 years old and 15–19 years old [44]. Its EFS and OS rates are both greater than 80% [45]. Lymphoblastic lymphoma most commonly arises from T lymphoblasts, but 20% of cases arise from B-cell lymphoblasts. Lymphoblastic lymphoma affects boys twice as often as girls [46]. Nearly a quarter of T-cell lymphoblastic lymphoma cases affect the mediastinum, and the disease can also be found to affect the bone marrow (BM); by comparison the disease is distinguished from T-cell leukemia considering that T-cell lymphoblastic lymphoma does not have more than 25% of BM involvement [47]. Moreover, recent genetic testing has revealed that mutation of NOTCH1/FBXW1 genes may reveal a disease that portends a good prognosis and gene mutations may allow risk stratification to occur as well as help elucidate T-cell lymphoblastic lymphoma as more distinct than T-cell leukemia [48,49]. More than 90% of T-cell lymphoblastic lymphomas express terminal deoxynucleotidyl transferase, CD2, CD3, and CD10, and about 20% express CD34 [47]. In a study conducted by the COG, CD33 expression and bone marrow involvement did not appear to significantly affect EFS, which was 86%; however, increasing bone marrow involvement did affect the 4-year OS rate, which was 88% in patients without bone marrow involvement but about 75% in those with bone marrow involvement [47].

3.5. Pediatric Follicular Lymphoma

Pediatric follicular lymphoma (PFL) is a rare NHL that is characterized by local lymphadenopathy and stage I/II disease. It can generally be cured by conservative management, such as surgical resection, without systemic chemotherapy or radiation. Unlike its adult counterpart, PFL is not known to recur after therapy [50,51,52]. In adults, follicular lymphoma makes up more than 20% of NHL cases, making it the second most common type of NHL, but in children and adolescents, PFL accounts for fewer than 2% of NHLs [52]. Owing to the rarity of PFL, the literature lacks detailed treatment guidelines or histopathologic characteristics [51]. Despite the lack of standard treatment, the EFS rate for patients with PFL is about 90% [53]. Unlike adult follicular lymphoma, in which 75–80% of patients harbor t(14;18), PFL is characterized by a lack of BCL2 rearrangement or over-expression of the bcl-2 protein [53,54].

3.6. Anaplastic Large-Cell Lymphoma

Anaplastic large cell lymphoma (ALCL) is a T-cell lymphoma that is rare in children and adolescents, with a median age of presentation of 16 years old, making up about 10% of NHLs in this population [44]. The disease generally presents in advanced stages (III-IV) with abdominal and mediastinal lymph node involvement as well as B-symptoms [55]. While BM disease is not common (<15% of cases), extra nodal spread to organs such as the skin, liver, and lung may be seen [56]. ALCL is characterized by large cells that express CD30; studies show that about 80% of cases harbor t(2;5), which encodes an NPM-ALK fusion protein. In murine hematopoietic stem cells, the introduction of this protein leads to malignant transformation [57,58]. In pediatric patients with ALCL, the overexpression of certain cytokines such as IL-6 and IL-2R also indicates that these patients have a distinct cytokine profile, although it is unclear if these makers have prognostic significance or signify a higher risk of developing ALCL [59]. There are no standard treatment guidelines [11,58]. However, ALK-positive disease appears to confer a better response to chemotherapy compared to ALK-negative ALCL [60]. In the pediatric population, patients with ALK-positive disease that involves the bone marrow have been found to have a 5-year PFS rate of 41% while those without bone marrow involvement have a 5-year PFS rate of 100%, indicating that bone marrow evaluation during the time of diagnosis may identify those that at risk of R/R disease [61].
The molecular characteristics and genetic abnormalities of the various pediatric and adolescent NHL types are described in Table 1.

4. Conventional Treatment of NHL in Children and Adolescents

The treatment of pediatric and adolescent NHL mainly consists of multidrug chemotherapy; radiation is not generally used in newly diagnosed or low-grade cases [3]. Moreover, unlike adult NHLs, certain pediatric NHLs, such as Burkitt lymphoma and DLBCL, share the same treatment backbone [7]. The treatment backbone of each pediatric and adolescent non-Hodgkin lymphoma is summarized in Table 1.

4.1. Treatment of Burkitt Lymphoma and DLBCL

Prior to the addition of rituximab to the systemic chemotherapy regimen for Burkitt lymphoma, treatment generally consisted of three phases: reduction, induction, and consolidation. The regimen is based on the FAB LMB 96 international study, which used a lower dose of cyclophosphamide than did prior trials [76]. The treatment regimens are further detailed in Table 1. Patte et al. [76] demonstrated that in patients with intermediate-risk Burkitt lymphoma or DLBCL which they defined as stage I/II that was not resected or stage III/IV without CNS involvement, the 4-year EFS rate was 90% and the OS rate was nearly 93%. That study indicated that a subgroup of pediatric and adolescent patients with intermediate-risk Burkitt lymphoma or DLBCL can be treated with 4 courses of a multidrug regimen, allowing the use of reduced doses of cyclophosphamide (3.3 g/m2) and doxorubicin (120 mg/m2) [76]. In those with high risk DLBCL or Burkitt lymphoma, defined by stage III disease with elevated lactate dehydrogenase (LDH) levels or stage IV disease, the risk of CNS disease is elevated along with the risk of relapse. However, we discuss below a phase II trial that shows the addition of rituximab to the LMB backbone for the treatment of upfront, high-grade, high-risk DLBCL and BL has been found to be associated with improved EFS rates compared to a group receiving chemotherapy only [77]. We also discuss improvements in prophylactic management of CNS disease associated with high-risk disease.

4.2. Treatment of PMLBCL

While there is no standard treatment for PMLBCL, the most common regimen used in the United States is DA-EPOCH-R (etoposide, doxorubicin, cyclophosphamide, and vincristine with rituximab) [41]. Moreover, whereas the adult regimen may include consolidative radiation because PMLBCL is radiosensitive, radiation therapy is generally not considered a part of upfront treatment in pediatric patients. Indeed, large studies of pediatric patients treated with radiation did not clearly establish a response to or a role for radiation [41,78]. In a 2019 study by Giulino-Roth et al. [78] of adult and pediatric patients with PMLBCL, patients who received DA-EPOCH-R had a 3-year EFS rate of nearly 86% and an OS rate of 95%; however, more children than adults developed thrombosis (46% vs. 23%, respectively). In contrast, a more recent phase 2 trial by Burke et al. [79] involving only pediatric (<18 years old) patients with PMLBCL demonstrated that treatment with DA-EPOCH-R did not confer better EFS or OS (70% and 85%, respectively) compared to control treatment which had similar rates. This international study also compared its results with historical survival rates from the FAB/LMB 96 study. In that study, patients with stage III MLBCL had 5-year EFS and OS rates of 66% and 73%, respectively, with a regimen consisting of pre-induction low-dose cyclophosphamide, vincristine, and prednisone followed by an induction course of COPADM (cyclophosphamide, prednisone, doxorubicin, and methotrexate) and a consolidation course of CYM (cytarabine and high-dose methotrexate) [34]. In the Berlin–Frankfurt–Munster (BFM) group study, pediatric patients received a pretreatment course of steroids and cyclophosphamide, followed by a regimen consisting of steroids, high-dose methotrexate, cytarabine, etoposide, doxorubicin, and cyclophosphamide or ifosfamide, with dose intensity based on tumor size and staging based on serum lactate dehydrogenase levels. The study found a 5-year EFS rate of 70%. Lactate dehydrogenase levels were found to have prognostic significance; patients with levels greater than or equal to 500 U/L had a higher risk of treatment failure, with an EFS rate of 40% [80]. Considering that multiple trials have demonstrated no significant differences in EFS and/or OS among regimens, there is still a need to find the optimal treatment that can produce EFS and OS rates comparable to those seen in other NHLs.

4.3. Treatment of ALCL

While there are no standard treatment guidelines for ALCL, these tumors are sensitive to a variety of chemotherapeutic regimens which are detailed in Table 1 [81]. For instance, in a study of patients enrolled in the NHL-BFM group trials, pediatric patients with ALK-positive ALCL had a 9-year EFS rate of 83% after receiving a short-term (5–7 month) chemotherapy regimen [66]. The French Society of Pediatric Oncology also conducted a study in pediatric ALCL in which patients received 2 courses of COPADM followed by a maintenance phase. This study found 3-year OS and EFS rates of 83% and 66%, respectively [82]. A COG study (ANHL0131) demonstrated that substituting vinblastine for vincristine in the standard consolidation regimen of doxorubicin, prednisone, and vincristine did not significantly affect EFS or OS rates. Patients who received vinblastine had EFS and OS rates of 79% and 87%, respectively, compared to the standard-therapy group’s rates of 80% and 91%, respectively [83].

4.4. Treatment of Lymphoblastic Lymphoma

The NHL-BFM 90 study demonstrated that patients with T-cell lymphoblastic lymphoma who are treated with a chemotherapy regimen similar to that used to treat T-cell acute lymphoblastic leukemia (T-ALL)—anthracycline and cyclophosphamide with no local radiation—had an excellent 5-year EFS rate of 90% [84,85]. One of the largest trials involving pediatric and adolescent patients with T-cell lymphoblastic lymphoma, COG AALL0434, demonstrated 4-year EFS and OS rates of 87% and 90%, respectively, in patients who received an augmented BFM regimen as detailed in Table 1 [69,70,71]. As further described below, whether introducing nelarabine to the chemotherapy backbone improves survival in T-cell lymphoblastic lymphoma has not yet been established, although in patients with T-ALL, nelarabine reduces the rate of CNS relapse. It has been found to be tolerable in patients with T-cell lymphoblastic lymphoma; neurological adverse events were the most common issue [69,85].

4.5. Treatment of PFL

As mentioned previously, PFL lacks a standard treatment regimen; however, good responses to treatment regimens as detailed in Table 1 [52]. Another study showed that treatment with R-CHOP was also effective and led to remission in 83% of patients aged 0–18 years over a median follow-up time of 31 months [86].

4.6. Treatment of R/R NHL

Although primary Burkitt lymphoma and DLBCL have 5-year EFS rates of nearly 90% in the pediatric population, patients with R/R NHL have substantially worse outcomes, with a cure rate of about 30% [87,88,89]. Among pediatric patients with R/R NHLs, those with PMLBCL have the best chance of survival; a study by Burkhardt et al. [89] evaluated over 600 patients and found 8-year survival rates of 57% for PMLBCL, 28% for Burkitt lymphoma, and 50% for DLBCL. In the same study, patients with DLBCL who did not undergo HSCT did not survive at 8 years. In the French LMB study of 773 pediatric patients, 4% of patients experienced relapse, mostly patients with Burkitt lymphoma [90]. In this study, the responses to first-line salvage chemotherapy with R-CYVE (rituximab, cytarabine, etoposide, and dexamethasone) and R-ICE (rituximab, ifosfamide, cyclophosphamide, and etoposide) were compared. The CR rate in patients receiving R-CYVE was 56%, compared to 43% in patients who received R-ICE [90]. Moreover, 85% of patients in the LMB study received rituximab alongside multiagent chemotherapy, but only about 30% of these patients survived, raising the question of the efficacy of rituximab. Overall, the R-ICE regimen appears to be the preferred salvage chemotherapy regimen, especially as studies have shown overall response rates of 60–70% [91,92].
Both allogeneic and autologous HSCT are options for pediatric patients with R/R NHL and appear to have similar outcomes [87]. A study by Rigaud et al. [90] detected no difference in 5-year survival rates between patients who underwent allogeneic or autologous transplants (50% and 54%, respectively). In a retrospective registry analysis by Gross et al. [93] of 182 pediatric patients with R/R NHL who underwent allogeneic or autologous HSCT, 5-year EFS rates were similar within disease types. In patients who received allogeneic or autologous HSCT for DLBCL, EFS rates were 50% and 52%, respectively. For patients with R/R Burkitt lymphoma, the EFS rates were 31% for allogeneic HSCT and 27% for autologous HSCT. Patients with R/R ALCL, too, had similar EFS rates after allogeneic or autologous HSCT (46% and 35%, respectively). In contrast, patients with lymphoblastic lymphoma had better outcomes after allogeneic HSCT, with a 5-year EFS rate of 40%, compared to 4% in patients who received autologous transplants. Outside of the results in patients with lymphoblastic lymphoma, the results are similar to those of a 10-year study by Giulino-Roth et al. [94] which found no difference in disease-free survival (DFS) in patients who underwent autologous or allogeneic HSCT, with DFS rates of 53% in both patient cohorts. However, outcomes were markedly different among disease types: patients with ALCL had the best outcomes, with 77% DFS and 83% OS, while the five included patients with Burkitt lymphoma all died 1–3 months following HSCT. Taken together, these studies demonstrate that HSCT, regardless of transplant source, represents a chance for cure with patients with R/R ALCL and DLBCL (OS rates of about 70% and 60%, respectively) but less so for patients with Burkitt lymphoma (OS of about 30%) [89,95,96,97]. In contrast, in pediatric and adolescent patients that relapse or fail to respond following first-line chemotherapy and do not progress to HSCT, overall survival is significantly lower at less than 25% underscoring the importance of HSCT to provide greater chance of survival [98].
Although autologous and allogeneic HSCT have similar EFS and OS rates, some researchers posit that allogeneic transplantation may induce a graft-versus-lymphoma (GVL) effect. In theory, withdrawal of the immunosuppressive agents typically given following HSCT or use of nonmyeloablative preparation regimens in those with minimal disease prior to transplantation would allow engraftment and cause the host’s antigen-presenting cells to present antigens to donor T cells, which then produce a GVL response [99,100]. Bishop et al. [101] demonstrated, in patients with DLBLCL that had relapsed or were not in CR by day 100 after HSCT, that withdrawal of immunosuppression or infusion of donor lymphocytes led to at least partial remission in 60% of patients, with patients remaining alive for at least 42 months. More than half of patients with a response also developed graft-versus-host disease (GVHD). In another recent study by Jeon et al. [102], patients with various forms of NHL who experienced chronic GVHD of greater than mild severity had better OS rates than did those with acute GVHD, indicating that chronic GVHD may lead to a GVL effect. These results demonstrate that a GVL effect can be induced to target NHL tumors following allogeneic HSCT. Reduced-intensity conditioning regimens administered before allogeneic HSCT have also been used to induce a GVL effect. In a mixed leukemia–lymphoma study of over 1000 adult patients, Storb et al. [103] demonstrated that patients with any stage of NHL who received reduced-intensity conditioning with total body irradiation with or without fludarabine had the lowest risk of relapse among all comparison groups. These data indicate that reduced-intensity conditioning not only reduces side effects associated with preparative regimens prior to HSCT but also allows engraftment that can sustain the GVL effect.

5. Advances in the Treatment of Pediatric and Adolescent NHL

While conventional chemotherapy has positive results in pediatric and adolescent patients with NHL, especially in those with Burkitt lymphoma or DLBCL, the outcomes of patients with R/R NHL are dismal, even when HSCT is used. Moreover, chemotherapy, including preconditioning chemotherapy prior to HSCT, carries serious adverse effects such as mucositis, decreased appetite, nausea and vomiting, and susceptibility to infections, which can hinder patients’ recovery [104]. Discussed below are promising new drugs and immunotherapies for the treatment of NHL including R/R NHL. We also discuss areas where there is a need for further research into the application of these treatment modalities for pediatric or adolescent patients with NHL. These modalities are further summarized in Table 2.

5.1. Targeting Cell-Surface Molecules and DNA Replication

Rituximab is a monoclonal antibody that binds the CD20 receptors on lymphoma cells, leading to antibody-dependent and complement-mediated cytotoxicity. It has produced favorable results in studies of adults with high-risk lymphomas [143]. In a 2020 study, Minard-Colin et al. [77] analyzed the effects of adding rituximab to the LMB chemotherapy backbone, mainly for patients with Burkitt lymphoma and DLBCL. Pediatric patients with high-risk disease who received rituximab plus LMB had a significantly higher (one sided p-value of 0.00096) EFS rate than did similar patients who received LMB alone (93.9% vs. 82.3%, respectively). In addition, a COG pilot study showed that rituximab is safe when combined with standard chemotherapy and resulted in 3-year EFS rates of 95% for patients with intermediate-risk FAB group B (stage III/IV) disease and 90% for those with high-risk FAB group C disease [104,144]. Outcomes were comparable for patients with and without CNS involvement [104,145,146]. Moreover, the use of high-dose methotrexate for early CNS prophylaxis with an alternating regimen of R-CHOP and R-CHOP plus etoposide has not only shown improved failure-free-survival (FFS) rates of 74%, but a CNS progression rate of 2.3%, a decrease from 5% in regimens involving R-CHOP only [147]. Most recently, the European Intergroup/COG Inter-B-NHL Ritux 2010 trial, which randomized high-risk pediatric patients with Burkitt lymphoma or DLBCL to chemotherapy with or without rituximab, was terminated early because it found a substantial benefit in the rituximab arm, with a 1-year EFS rate of 94.2% compared to 81.5% in the control arm [77,104]. Further, in a study of pediatric patients with advanced (stage III/IV) NHL, Goldman et al. [148] demonstrated that the use of rituximab was associated with a 60% reduction in the anthracycline dose needed to maintain acceptable EFS and OS rates. Indeed, the researchers showed that rituximab may permit reduction of anthracycline dosing to 50 mg/m2 total, in light of the 100% EFS and OS rates at 4 years in the FAB group B arm. Taken together, these studies indicate that rituximab has great utility for treating CD20-positive NHL, including advanced-stage disease, and can permit reduction of conventional chemotherapy dosages to reduce adverse effects. In December 2021, the FDA approved rituximab for use in combination with chemotherapy in patients between the ages of 6 months and 18 years with DLBCL or Burkitt lymphoma on the basis of the results of the Inter-B-NHL-Ritux 2010 trial [77,149].
Brentuximab vedotin (BV) is a CD30-directed antibody conjugated to the microtubule-disrupting agent monomethyl auristatin E and works to induce apoptosis in CD30-positive cells during mitosis [150]. Because it targets receptors on lymphoma cells, BV has the added benefit of fewer off-target toxic effects than systemic chemotherapy [151]. In a phase 1/2 study of BV in pediatric patients with classical Hodgkin lymphoma and ALCL, 77% of patients had a reduction in tumor size, and the OS rate at 12 months was 79%; moreover, the safety profile was no worse than that observed in adults [151]. In the COG trial ANHL12P1, which included pediatric patients with ALCL, the addition of BV to conventional chemotherapy led to no additional toxic effects, including no grade 3 or 4 neuropathy and 2-year OS was 97% and 2-year EFS was 79%, indicating that BV may be useful when added to chemotherapy regimens for pediatric patients with ALCL [109]. BV also shows promise as an adjunct agent to help induce remission in patients with ALCL and as a bridging immunotherapy while patients await HSCT.
Blinatumomab is a bispecific T-cell-engaging antibody that targets the CD19 receptor on leukemia cells. When used in the postinduction phase for treatment of relapsed B-cell ALL in pediatric patients, it has been shown to improve EFS and OS rates [152,153]. Blinatumomab links the targeting regions of two antibodies directed against CD19 and CD3. CD19 is expressed by precursor-B-ALL cells, and CD3 is a constant part of the T-cell receptor complex [152,153]. While blinatumomab has been associated with acute side effects such as cytokine release syndrome (CRS) and neurotoxicity, there do not appear to be significant long-term neurotoxic effects in adult patients with R/R NHL treated with blinatumomab [154]. While the use of blinatumomab has not been approved for pediatric patients with NHL, phase 1 and 2 trials have shown that blinatumomab has great promise. Goebler et al. [155,156] demonstrated overall response rates (ORRs) of nearly 70% for patients with all NHLs and 55% for patients with DLBCL with a dose of 60 µg/m2 per day serving as the maximum tolerated dose. Dufner et al. [154] found similar results; in their study, use of blinatumomab at a maximum tolerated dose of 60 or more µg/m2 per day led to a median OS duration of nearly 8 years in patients with R/R NHL, with minimal to no long-term adverse effects. In contrast, patients treated at a dose below 60 µg/m2 per day had a median OS of just over 1 year. Considering that early phase trials have shown that blinatumomab has some activity in adults with R/R NHL, further prospective and retrospective studies are needed to determine the maximum tolerated dose in the pediatric NHL population and the impact of blinatumomab treatment on EFS and OS, especially in patients with R/R NHL [154,157].
Another cell surface effector, Inotuzumab ozogamicin is a CD22-directed antibody that is conjugated with calicheamicin, a cytotoxin that leads to the death of CD22-expressing leukemia cells. In several clinical trials, inotuzumab ozogamicin has been shown to induce remission of R/R ALL with minimal off-target effects owing to its specificity for the CD22 receptor on leukemia cells [158]. While phase 2 studies of inotuzumab ozogamicin are currently ongoing for adults with R/R NHL, including those whose tumors are not responsive to rituximab, no data has been published for the use of inotuzumab in pediatric and adolescent population with NHL. Thus, this is an area in which further research focused on pediatric NHL is required especially considering that the role of inotuzumab in the treatment of R/R pediatric ALL has been positive in ongoing early phase trials from the Children’s Oncology Group (Clinicaltrials.gov identifier NCT02981628) and a European-based multi-institutional study, ITCC-059, which shows an overall response rate of over 80% in patients with R/R CD22+ ALL who were treated with inotuzumab. Considering that these studies are enrolling patients with B-cell lymphoid malignancies, including NHL, there is a possibility that inotuzumab may be associated with positive outcomes in the treatment of CD22+ R/R NHL [104,120,159]. However, it must be noted that inotuzumab has been associated with increased risk of veno-occlusive disease (VOD) in those patients subsequently receiving an allogenic transplant, which can be fatal [160].
Glofitamab and mosunetuzumab have also emerged as bi-specific antibodies showing promise in the treatment of R/R NHL [161]. Although currently in early phase clinical trials, glofitamab, a bi-specific antibody that binds the CD3 receptors on T cells and CD20 receptors on B cells, has been studied in combination with R-CHOP with great response. A phase IB clinical trial has shown that the combination regimen induces an OR of nearly 90% at 9 months in patients with R/R NHL and a CR of 100% in patients in patients with untreated DLBCL [162]. Moreover, CRS rates were found to be significantly low with 3% of patients experiencing ≥ grade 3 CRS in the R/R NHL group and no patients experiencing CRS in the upfront DLBCL group. Mosunetuzumab, a similar CD3-CD20 bi-specific engager, when used as a single agent has shown an objective response rate of 35% in those with aggressive, R/R NHL and an objective response rate of 66% in those with indolent, R/R NHL [163]. Similar to glofitamab, the CRS rate of those experiencing ≥ grade 3 CRS was low with a rate slightly greater than 1%. Considering that these results are from a phase I trial, more data is needed to determine the role of mosunetuzumab as a single agent or in combination with conventional chemotherapy in those with initial or R/R disease.
Nelarabine is a purine analog and a form of deoxyguanosine that inhibits DNA synthesis in T lymphoblasts [164]. Initially approved in 2005 for the treatment of R/R T-cell lymphoblastic lymphoma in pediatric and adult patients, the drug has been shown to be tolerated well by pediatric patients. However, despite earlier reports indicating the utility of nelarabine monotherapy for R/R T-cell lymphoblastic lymphoma with response rates of nearly 40%, the COG AALL0434 study did not find significant differences in outcomes when nelarabine was added as an adjunct to the standard chemotherapy regimen [165,166,167,168]. These data are intriguing because nelarabine has shown an OR rate as high as 97% in studies of adults with T-cell lymphoblastic lymphoma when used with combination chemotherapy. Thus, further research into the optimization of nelarabine use in the pediatric and adolescent population is required [169].

5.2. Cellular Therapy

CAR T-cell therapy has produced good results in pediatric R/R leukemia but has not been extensively studied in the pediatric NHL population [170]. CAR T-cell therapy relies on the modification of a patient’s T lymphocytes in order to express a cell surface receptor, most commonly for CD19, that can recognize leukemia cells expressing the CD19 surface antigen and thereby lead to on-target cytotoxicity with minimal off-target effects [171]. While four different anti-CD19 CAR T-cell products have been approved for use in adults, none have been approved yet for pediatric patients with NHL [172]. Various CAR T-cell products such as tisagenlecleucel, KTE-X19, and axicabtagene clioleucel have been tested in phase 2 and 3 trials in adult patients with R/R LBCL and have shown ORRs ranging from 50 to 80% and complete response rates (CRRs) of 40–50%, in some cases indicating improvements in response over other salvage treatment options [173,174,175,176]. These same studies also demonstrated that the most common adverse effects such as anemia and neutropenia were similar to those seen in standard of care salvage therapies and grade 3 CRS and neurotoxicity, known adverse effects of CAR T-cell therapy, were often manageable with little to no associated mortality. Curiously, the BELINDA phase 3 trial is the most recent study on the use of tisagenlecleucel in patients with high-risk R/R LBCL which indicates that it does not have better outcomes than standard salvage therapy and the CAR T-cell product was not superior to the standard product [177]. In the upfront setting, the ZUMA-12 study, a phase 2 trial in adult patients with high-risk LBCL who received axicabtagene cliolecuel following 2 cycles of either R-CHOP or DA-EPOCH-R, has demonstrated that axicabtagene clioleucel may have an effective role as a first-line therapy considering that patients demonstrated an ORR greater than 80% with tolerable CRS and neurotoxicity [178].
Zhang et al. [179] have also published promising preliminary results of a clinical trial in which pediatric patients with Burkitt lymphoma, DLBCL, and B-cell lymphoblastic lymphoma underwent multiple rounds of anti-CD19, anti-CD20, or anti-CD22 CAR T-cell therapy, with an ORR of 94% and a CRR of 71%. Of note, 53% of patients experienced severe cytokine release syndrome, and 41% experienced some form of neurotoxicity including seizures. At this time, the BIANCA trial (NCT0310724), a clinical trial to determine the safety and efficacy of tisagenlecleucel, an anti-CD19 CAR T-cell therapy, is underway and includes pediatric, adolescent, and young adult patients with R/R B-cell NHL such as Burkitt lymphoma, DLBCL, and PMLBCL. Thus far, adverse effects have been tolerable, with no patients having cytokine release syndrome greater than grade 3 in severity [180]. CAR T-cell therapy is thus a potential treatment modality for patients with R/R NHL on the basis of its promising preliminary results in pediatric patients and its positive outcomes in adult patients with NHL [181].
While no large trials have yet evaluated the use of axicabtagene ciloleucel in pediatric patients with NHL, a case report of a 19-year-old with PMLBCL receiving axicabtagene ciloleucel indicated positive results with manageable adverse effects such as immune-effector cell-associated neurotoxicity syndrome (ICANS) [182]. These results indicate that axicabtagene ciloleucel is not only effective in inducing a response in R/R large B-cell lymphoma, but the response is sustained and possibly superior to the standard of care regimen of salvage chemotherapy followed, in those patients who respond, by high dose chemotherapy followed by stem cell rescue in patients with R/R disease. Taking into account the positive results of the ZUMA-7 trial, axicabtagene ciloleucel for the treatment of R/R LBCL was approved by the FDA in April 2022, joining tisagenlecleucel and lisocabtagene maraleucel as other anti-CD19 CAR T-cell products approved by the FDA for the treatment of R/R LBCL 177, [183,184,185]. The latter was approved in February 2021 after the phase 1 TRANSCEND NHL 001 trial indicated an ORR of over 70% and a CRR over 50%, with grade 3 or 4 CRS occurring in 1% of patients and grade 3 or 4 neurotoxicity in 10% of patients [184].
The phase 1/2 ZUMA-4 trial is underway to evaluate the anti-CD19 CAR T-cell therapy, KTE-X19, in the treatment of patients aged 21 years and under with DLBCL, PMLBCL, and Burkitt lymphoma that is refractory to or has failed two lines of therapy [186]. Preliminary results of the phase 1 portion of ZUMA-4 in patients with ALL are promising, with complete remission rates ranging from 64% to 75% across the various dosing groups. Moreover, 100% of patients with responses had negative minimal residual disease [187]. Thus, the preliminary outcomes of these trials indicate a rationale for the expansion of CAR T-cell products for the treatment of R/R pediatric NHL.
In patients in whom anti-CD19 CAR T-cell therapy fails, targeting CD22 may be an alternative. A current clinical trial has demonstrated that in patients whose large B-cell lymphoma has relapsed after CD19 CAR T-cell therapy, infusion of CD22-directed CAR T cells is safe and can lead to a complete remission lasting as long as 8 months [188]. While the most common CAR T-cell products singly target the CD19 receptor on cancer cells, researchers have also engineered dual-targeting CAR T-cells that recognize both CD19 and CD22 receptors on lymphoma cells. A recent study by Hu et al. [189] evaluated the effect of dual CD19/CD22 CAR T-cell products on R/R B-cell lymphoma; over 60% of patients had a CR, with a 2-year OS rate of 77% and PFS rate of 40%. In contrast, a phase 1 trial found that the use of dual anti-CD19/CD22 CAR T-cell therapy led to a 6-month PFS rate similar to that of tisagenlecleucel (29%), leading to closure of the large B-cell lymphoma arm [190]. Considering that response rates are generally worse in adults with large B-cell lymphoma than in children and that these studies primarily enrolled adult patients, extrapolation to the pediatric and adolescent population may be difficult, indicating that further trials are needed to determine the response of dual-targeting CAR T cells against pediatric NHL. Compounds which improve CAR T-cell killing such as decitabine may also serve as supplemental treatments to ensure long-term killing of CAR T-cells in order to avoid exhaustion and subsequent relapse [191,192]. Indeed, multiple early phase clinical trials exist to determine the use of decitabine primed CAR T-cells in their role in treating R/R NHL (clinicaltrial.gov identifiers, NCT04697940, NCT04850560) [193].
Another form of cellular therapy involves the use of antigen-specific T-cells directed towards other targets on NHL cells. One such modality that has shown promising results is EBV-specific T-cells from an autologous source, which target the latent membrane protein on EBV-associated NHL cells [194,195]. A study by Bollard et al. [195] included pediatric and adolescent patients and showed that infusion of cytotoxic T lymphocytes that target the latent membrane proteins on EBV-associated NHL or Hodgkin lymphoma cells produced durable remissions in more than 80% of patients at 2 years following infusion. Moreover, no patients experienced infusion-related toxicities.
Natural killer (NK) cells can recognize and kill tumor cells with minimal toxicity, as demonstrated in studies where NK cells have been infused alongside or in close timing with HSCT and demonstrated expansion and antileukemia effects, especially in acute myeloid leukemia [196]. While no large trials exclusively in pediatric and adolescent patients have determined the utility of NK cell infusion for the treatment of NHL, Liu et al. [197] have demonstrated in early-phase trials that NK cells engineered with an anti-CD19 CAR induced responses in four of six patients with R/R NHL at a median follow-up time of nearly 14 months. In addition, the NK cells persisted for at least 12 months following infusion. Moreover, a phase 2 clinical trial of patients older than 15 years with R/R NHL is currently studying the safety and 30-day mortality risk of a combination of NK cells derived from umbilical cord blood along with rituximab, chemotherapy, and HSCT; the study will also determine EFS, OS, and the persistence of the cord blood-derived NK cells [198]. Thus, NK cells are an exciting modality for the treatment of R/R NHL with minimal off-site toxicities.
Donor lymphocyte infusion (DLI) is another example of a modality that can induce a GVL effect in patients with NHL. In a 2018 retrospective registry analysis by Robinson et al. [199], adult patients with NHL who experienced relapse after allogeneic HSCT and then received DLI had a median response duration of 36 months; patients with follicular lymphoma had the highest OR rate among the NHL subtypes, 84%, with nearly 70% having CR [199]. While no large studies have yet determined the optimal use of DLI in pediatric patients with NHL, Gozdzik et al. [133] found that DLI in pediatric patients following HSCT, including those with Hodgkin lymphoma and NHL, led to a CR rate of 35%, although response rates were not broken down by cancer type. Moreover, 4% of patients died of DLI-induced GVHD. There is an obvious need for further study of the role of DLI in the treatment of pediatric NHLs in order to determine its efficacy, safety, and short- and long-term complications, including GVHD.
Tumor-infiltrating lymphocytes (TILs) are similar to NK cells in that they are an autologous source of cellular therapy that can be engineered to target certain tumor cells [200]. Researchers have demonstrated that administration of TILs with cyclophosphamide for the treatment of metastatic melanoma has a response rate of nearly 50%, with complete responses lasting more than 20 months and partial responses lasting a median of 4 months [201]. While studies have not determined the response of NHL to TIL therapy, researchers have shown that TILs can be isolated and engineered from lymphomatous tumors such DLBCL and follicular lymphoma, indicating that they may have future utility in the treatment of NHL [202].

5.3. Immune Checkpoint Inhibitors

In June 2018, the FDA approved the use of pembrolizumab for the treatment of adult and pediatric R/R PMLBCL [203]. Pembrolizumab is a small-molecule inhibitor targeting programmed cell death protein-1 (PD-1) on lymphocytes. This prevents tumor-secreted programmed cell death protein ligand-1 (PD-L1) from attaching and leading to lymphocyte deactivation and tumor evasion of immune system detection [204]. In the KEYNOTE trial, patients with R/R PMLBCL who received pembrolizumab tolerated the drug well, with none experiencing grade 5 adverse events and nearly 50% showing an overall response [205]. Current trials are also underway in testing the concurrent use of pembrolizumab with a host of other drugs, including rituximab and other immunotherapies such as CAR T-cell therapy, in order to improve outcomes in patients with R/R PMLBCL [203].
Phase 1/2 trials of another PD-1 inhibitor, nivolumab, showed safety and efficacy in pediatric patients with solid tumors and R/R classical Hodgkin lymphoma; however, its efficacy in the treatment of aggressive NHL remains unclear [137]. In the adult population, nivolumab has shown favorable tolerability, but the ORR is dismal in those with R/R disease, as evidenced by objective response rates of 10% in patients whose disease did not respond to autologous HSCT and 3% in patients who did not receive autologous HSCT [206]. Atezolizumab, a PD-L1 inhibitor, was also well tolerated in a large phase 1/2 study in pediatric patients with solid tumors, classical Hodgkin lymphoma, and NHL but its responses were also suboptimal, as only 5% of patients had a partial response at 6 months [207].

5.4. Targeted Therapy

As discussed above, nearly 90% of patients with ALCL harbor the ALK-NPM fusion protein on tumor cells, which can serve as a target for immunotherapy [208]. One such agent is crizotinib, a tyrosine kinase inhibitor that targets the ALK-ROS1-MET pathway in non-small cell lung cancer [209]. In a COG study, crizotinib monotherapy showed response rates of 80–90%, indicating that for patients with unresectable ALCL, crizotinib may be an acceptable form of immunotherapy and a bridge to HSCT [210]. On the basis of results from the COG ADVL0912 trial, in which nearly 40% of patients had a response lasting for up to 6 months, in January 2021 the FDA approved crizotinib for use in pediatric patients with R/R ALCL [211].
JAK2 inhibition may be useful in PMLBCL, which has been found to have mutations in pathways related to JAK/STAT signaling. In preclinical mouse xenograft models, JAK2 inhibition led to decreased tumor growth and longer survival in models of MLBCL and Hodgkin lymphoma [212]. In contrast, a small-scale study of patients greater than 19 years old detailed the effectiveness of Ruxolitinib, a JAK/STAT inhibitor, in the treatment of Hodgkin lymphoma but there was no response in PMLBCL. These studies underscore the need for larger trials that study the use of Ruxolitinib in the treatment of pediatric and adolescent PMBLCL or other NHL that may show amplification of the JAK pathway [213].

6. Conclusions

NHL in pediatric and adolescent patients is distinct type from the NHLs seen in adults. While the outcomes of pediatric and adolescent patients with primary NHLs such Burkitt lymphoma and DLBCL are outstanding, survival rates markedly decrease following relapse [89]. HSCT provides some hope for patients with R/R disease, although the results depend on the type of pediatric NHL. Here, we highlighted areas of recent progress in the treatment of NHL. We emphasize that recent advances are characterized by targeting treatment to cell-surface markers and cell receptors deployed by the tumor to evade detection by the immune system. While the current evolution in treatment is promising, further research is needed in areas such as the use of CAR T-cell therapy and monoclonal antibodies, especially as these treatments have had promising safety and response profiles in other pediatric cancers such as leukemia.

Author Contributions

Conceptualization, I.N.S. and S.K.; writing—original draft preparation, I.N.S. and A.E.; writing—review and editing, A.E., D.R., K.M.M., Y.N. and S.K.; supervision, S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would also like to acknowledge Amy Ninetto of the University of Texas MD Anderson Cancer Center Research Medical Library for their assistance in the editing and revision of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bowzyk Al-Naeeb, A.; Ajithkumar, T.; Behan, S.; Hodson, D.J. Non-Hodgkin lymphoma. BMJ 2018, 362, k3204. [Google Scholar] [CrossRef] [PubMed]
  3. Sandlund, J.T. Should Adolescents with NHL Be Treated as Old Children or Young Adults? Hematology 2007, 2007, 297–303. [Google Scholar] [CrossRef]
  4. Ramirez, L.Y.; Huestis, S.E.; Yap, T.Y.; Zyzanski, S.; Drotar, D.; Kodish, E. Potential chemotherapy side effects: What do oncologists tell parents? Pediatr. Blood Cancer 2009, 52, 497–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Svoboda, J.; Bair, S.M.; Landsburg, D.J.; Nasta, S.D.; Nagle, S.J.; Barta, S.K.; Khan, N.; Filicko-O’Hara, J.; Gaballa, S.; Strelec, L.; et al. Brentuximab vedotin in combination with rituximab, cyclophosphamide, doxorubicin, and prednisone as frontline treatment for patients with CD30-positive B-cell lymphomas. Haematologica 2021, 106, 1705–1713. [Google Scholar] [CrossRef] [PubMed]
  6. Sandlund, J.T.; Martin, M.G. Non-Hodgkin lymphoma across the pediatric and adolescent and young adult age spectrum. Hematol. Am. Soc. Hematol. Educ. Prog. 2016, 2016, 589–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Dunleavy, K.; Gross, T.G. Management of aggressive B-cell NHLs in the AYA population: An adult vs pediatric perspective. Blood 2018, 132, 369–375. [Google Scholar] [CrossRef] [Green Version]
  8. Wood, W.A.; Lee, S.J. Malignant hematologic diseases in adolescents and young adults. Blood 2011, 117, 5803–5815. [Google Scholar] [CrossRef] [Green Version]
  9. Linabery, A.M.; Ross, J.A. Childhood and adolescent cancer survival in the US by race and ethnicity for the diagnostic period 1975–1999. Cancer 2008, 113, 2575–2596. [Google Scholar] [CrossRef] [Green Version]
  10. Tai, E.; Pollack, L.A.; Townsend, J.; Li, J.; Steele, C.B.; Richardson, L.C. Differences in Non-Hodgkin Lymphoma Survival between Young Adults and Children. Arch. Pediatr. Adolesc. Med. 2010, 164, 218–224. [Google Scholar] [CrossRef] [Green Version]
  11. Burkhardt, B.; Oschlies, I.; Klapper, W.; Zimmermann, M.; Woessmann, W.; Meinhardt, A.; Landmann, E.; Attarbaschi, A.; Niggli, F.; Schrappe, M.; et al. Non-Hodgkin’s lymphoma in adolescents: Experiences in 378 adolescent NHL patients treated according to pediatric NHL-BFM protocols. Leukemia 2011, 25, 153–160. [Google Scholar] [CrossRef] [PubMed]
  12. Hecht, J.L.; Aster, J.C. Molecular biology of Burkitt’s lymphoma. J. Clin. Oncol. 2000, 18, 3707–3721. [Google Scholar] [CrossRef]
  13. Molyneux, E.M.; Rochford, R.; Griffin, B.; Newton, R.; Jackson, G.; Menon, G.; Harrison, C.J.; Israels, T.; Bailey, S. Burkitt’s lymphoma. Lancet 2012, 379, 1234–1244. [Google Scholar] [CrossRef] [Green Version]
  14. Schmitz, R.; Ceribelli, M.; Pittaluga, S.; Wright, G.; Staudt, L.M. Oncogenic mechanisms in Burkitt lymphoma. Cold Spring Harb. Perspect. Med. 2014, 4, a014282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Boxer, L.M.; Dang, C.V. Translocations involving c-myc and c-myc function. Oncogene 2001, 20, 5595–5610. [Google Scholar] [CrossRef] [Green Version]
  16. Schulz, T.F.; Boshoff, C.H.; Weiss, R.A. HIV infection and neoplasia. Lancet 1996, 348, 587–591. [Google Scholar] [CrossRef]
  17. Graham, B.S.; Lynch, D.T. Burkitt Lymphoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  18. Epstein, M.A.; Achong, B.G.; Barr, Y.M. Virus Particles in Cultured Lymphoblasts from Burkitt’s Lymphoma. Lancet 1964, 1, 702–703. [Google Scholar] [CrossRef]
  19. Epstein, M.A.; Henle, G.; Achong, B.G.; Barr, Y.M. Morphological and Biological Studies on a Virus in Cultured Lymphoblasts from Burkitt’s Lymphoma. J. Exp. Med. 1965, 121, 761–770. [Google Scholar] [CrossRef] [Green Version]
  20. Brady, G.; MacArthur, G.J.; Farrell, P.J. Epstein-Barr virus and Burkitt lymphoma. J. Clin. Pathol. 2007, 60, 1397–1402. [Google Scholar] [CrossRef]
  21. Lavu, E.; Morewaya, J.; Maraka, R.; Kiromat, M.; Ripa, P.; Vince, J. Burkitt lymphoma in Papua New Guinea—40 Years on. Ann. Trop. Paediatr. 2005, 25, 191–197. [Google Scholar] [CrossRef]
  22. Parkin, D.; Ferlay, J.; Hamdi-Cherif, M.; Sitas, F.; Thomas, J.; Wabinga, H.; Whelan, S. Cancer in Africa. Epidemiol. Prev. 2003, 4, 268–276. [Google Scholar]
  23. Allen, C.E.; Kelly, K.M.; Bollard, C.M. Pediatric lymphomas and histiocytic disorders of childhood. Pediatr. Clin. N. Am. 2015, 62, 139–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. England, R.J.; Pillay, K.; Davidson, A.; Numanoglu, A.; Millar, A.J. Intussusception as a presenting feature of Burkitt lymphoma: Implications for management and outcome. Pediatr. Surg. Int. 2012, 28, 267–270. [Google Scholar] [CrossRef] [PubMed]
  25. Gupta, H.; Davidoff, A.M.; Pui, C.H.; Shochat, S.J.; Sandlund, J.T. Clinical implications and surgical management of intussusception in pediatric patients with Burkitt lymphoma. J. Pediatr. Surg. 2007, 42, 998–1001. [Google Scholar] [CrossRef]
  26. Padala, S.A.; Kallam, A. Diffuse Large B Cell Lymphoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  27. Woessmann, W.; Seidemann, K.; Mann, G.; Zimmermann, M.; Burkhardt, B.; Oschlies, I.; Ludwig, W.-D.; Klingebiel, T.; Graf, N.; Gruhn, B.; et al. The impact of the methotrexate administration schedule and dose in the treatment of children and adolescents with B-cell neoplasms: A report of the BFM Group Study NHL-BFM95. Blood 2005, 105, 948–958. [Google Scholar] [CrossRef] [Green Version]
  28. Rovira, J.; Valera, A.; Colomo, L.; Setoain, X.; Rodríguez, S.; Martínez-Trillos, A.; Giné, E.; Dlouhy, I.; Magnano, L.; Gaya, A.; et al. Prognosis of patients with diffuse large B cell lymphoma not reaching complete response or relapsing after frontline chemotherapy or immunochemotherapy. Ann. Hematol. 2015, 94, 803–812. [Google Scholar] [CrossRef] [Green Version]
  29. Nowakowski, G.S.; Czuczman, M.S. ABC, GCB, and Double-Hit Diffuse Large B-Cell Lymphoma: Does Subtype Make a Difference in Therapy Selection? Am. Soc. Clin. Oncol. Educ. Book 2015, 35, e449–e457. [Google Scholar] [CrossRef]
  30. Schuetz, J.M.; Johnson, N.A.; Morin, R.D.; Scott, D.W.; Tan, K.; Ben-Nierah, S.; Boyle, M.; Slack, G.W.; Marra, M.A.; Connors, J.M.; et al. BCL2 mutations in diffuse large B-cell lymphoma. Leukemia 2012, 26, 1383–1390. [Google Scholar] [CrossRef] [Green Version]
  31. Miles, R.R.; Raphael, M.; McCarthy, K.; Wotherspoon, A.; Lones, M.A.; Terrier-Lacombe, M.J.; Patte, C.; Gerrard, M.; Auperin, A.; Sposto, R.; et al. Pediatric diffuse large B-cell lymphoma demonstrates a high proliferation index, frequent c-Myc protein expression, and a high incidence of germinal center subtype: Report of the French-American-British (FAB) international study group. Pediatr. Blood Cancer 2008, 51, 369–374. [Google Scholar] [CrossRef] [Green Version]
  32. Matulis, S.M.; Boise, L.H. BCL2 dependency in diffuse large B-cell lymphoma: It’s a family affair. Haematologica 2020, 105, 1993–1996. [Google Scholar] [CrossRef]
  33. Oschlies, I.; Klapper, W.; Zimmermann, M.; Krams, M.; Wacker, H.-H.; Burkhardt, B.; Harder, L.; Siebert, R.; Reiter, A.; Parwaresch, R. Diffuse large B-cell lymphoma in pediatric patients belongs predominantly to the germinal-center type B-cell lymphomas: A clinicopathologic analysis of cases included in the German BFM (Berlin-Frankfurt-Münster) Multicenter Trial. Blood 2006, 107, 4047–4052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gerrard, M.; Waxman, I.M.; Sposto, R.; Auperin, A.; Perkins, S.L.; Goldman, S.; Harrison, L.; Pinkerton, R.; McCarthy, K.; Raphael, M.; et al. Outcome and pathologic classification of children and adolescents with mediastinal large B-cell lymphoma treated with FAB/LMB96 mature B-NHL therapy. Blood 2013, 121, 278–285. [Google Scholar] [CrossRef] [PubMed]
  35. Dabrowska-Iwanicka, A.; Walewski, J.A. Primary mediastinal large B-cell lymphoma. Curr. Hematol. Malig. Rep. 2014, 9, 273–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Attias, D.; Hodgson, D.; Weitzman, S. Primary mediastinal B-cell lymphoma in the pediatric patient: Can a rational approach to therapy be based on adult studies? Pediatr. Blood Cancer 2009, 52, 566–570. [Google Scholar] [CrossRef] [PubMed]
  37. Oschlies, I.; Burkhardt, B.; Salaverria, I.; Rosenwald, A.; d’Amore, E.S.G.; Szczepanowski, M.; Koch, K.; Hansmann, M.L.; Stein, H.; Möller, P.; et al. Clinical, pathological and genetic features of primary mediastinal large B-cell lymphomas and mediastinal gray zone lymphomas in children. Haematologica 2011, 96, 262–268. [Google Scholar] [CrossRef] [Green Version]
  38. Steidl, C.; Gascoyne, R.D. The molecular pathogenesis of primary mediastinal large B-cell lymphoma. Blood 2011, 118, 2659–2669. [Google Scholar] [CrossRef] [Green Version]
  39. Paulli, M.; Sträter, J.; Gianelli, U.; Rousset, M.T.; Gambacorta, M.; Orlandi, E.; Klersy, C.; Lavabre-Bertrand, T.; Morra, E.; Manegold, C.; et al. Mediastinal B-cell lymphoma: A study of its histomorphologic spectrum based on 109 cases. Hum. Pathol. 1999, 30, 178–187. [Google Scholar] [CrossRef]
  40. Reiter, A.; Klapper, W. Recent advances in the understanding and management of diffuse large B-cell lymphoma in children. Br. J. Haematol. 2008, 142, 329–347. [Google Scholar] [CrossRef]
  41. Giulino-Roth, L. How I treat primary mediastinal B-cell lymphoma. Blood 2018, 132, 782–790. [Google Scholar] [CrossRef]
  42. Yuan, J.; Wright, G.; Rosenwald, A.; Steidl, C.; Gascoyne, R.D.; Connors, J.M.; Mottok, A.; Weisenburger, D.D.; Greiner, T.C.; Fu, K.; et al. Identification of Primary Mediastinal Large B-cell Lymphoma at Nonmediastinal Sites by Gene Expression Profiling. Am. J. Surg. Pathol. 2015, 39, 1322–1330. [Google Scholar] [CrossRef]
  43. Lones, M.A.; Perkins, S.L.; Sposto, R.; Kadin, M.E.; Kjeldsberg, C.R.; Wilson, J.F.; Cairo, M.S. Large-Cell Lymphoma Arising in the Mediastinum in Children and Adolescents Is Associated with an Excellent Outcome: A Children’s Cancer Group Report. J. Clin. Oncol. 2000, 18, 3845–3853. [Google Scholar] [CrossRef] [PubMed]
  44. Derebas, J.; Panuciak, K.; Margas, M.; Zawitkowska, J.; Lejman, M. The New Treatment Methods for Non-Hodgkin Lymphoma in Pediatric Patients. Cancers 2022, 14, 1569. [Google Scholar] [CrossRef] [PubMed]
  45. Burkhardt, B.; Hermiston, M.L. Lymphoblastic lymphoma in children and adolescents: Review of current challenges and future opportunities. Br. J. Haematol. 2019, 185, 1158–1170. [Google Scholar] [CrossRef]
  46. Burkhardt, B.; Zimmermann, M.; Oschlies, I.; Niggli, F.; Mann, G.; Parwaresch, R.; Riehm, H.; Schrappe, M.; Reiter, A.; BFM Group. The impact of age and gender on biology, clinical features and treatment outcome of non-Hodgkin lymphoma in childhood and adolescence. Br. J. Haematol. 2005, 131, 39–49. [Google Scholar] [CrossRef]
  47. Patel, J.L.; Smith, L.M.; Anderson, J.; Abromowitch, M.; Campana, D.; Jacobsen, J.; Lones, M.A.; Gross, T.G.; Cairo, M.S.; Perkins, S.L. The immunophenotype of T-lymphoblastic lymphoma in children and adolescents: A Children’s Oncology Group report. Br. J. Haematol. 2012, 159, 454–461. [Google Scholar] [CrossRef]
  48. Burkhardt, B. Paediatric lymphoblastic T-cell leukaemia and lymphoma: One or two diseases? Br. J. Haematol. 2010, 149, 653–668. [Google Scholar] [CrossRef]
  49. Balbach, S.T.; Makarova, O.; Bonn, B.R.; Zimmermann, M.; Rohde, M.; Oschlies, I.; Klapper, W.; Rössig, C.; Burkhardt, B. Proposal of a genetic classifier for risk group stratification in pediatric T-cell lymphoblastic lymphoma reveals differences from adult T-cell lymphoblastic leukemia. Leukemia 2016, 30, 970–973. [Google Scholar] [CrossRef] [PubMed]
  50. Louissaint, A., Jr.; Ackerman, A.M.; Dias-Santagata, D.; Ferry, J.A.; Hochberg, E.P.; Huang, M.S.; Iafrate, A.J.; Lara, D.O.; Pinkus, G.S.; Salaverria, I.; et al. Pediatric-type nodal follicular lymphoma: An indolent clonal proliferation in children and adults with high proliferation index and no BCL2 rearrangement. Blood 2012, 120, 2395–2404. [Google Scholar] [CrossRef] [Green Version]
  51. Atra, A.; Meller, S.T.; Stevens, R.S.; Hobson, R.; Grundy, R.; Carter, R.L.; Pinkerton, C.R. Conservative management of follicular non-Hodgkin’s lymphoma in childhood. Br. J. Haematol. 1998, 103, 220–223. [Google Scholar] [CrossRef]
  52. Oschlies, I.; Salaverria, I.; Mahn, F.; Meinhardt, A.; Zimmermann, M.; Woessmann, W.; Burkhardt, B.; Gesk, S.; Krams, M.; Reiter, A.; et al. Pediatric follicular lymphoma—A clinico-pathological study of a population-based series of patients treated within the Non-Hodgkin’s Lymphoma—Berlin-Frankfurt-Munster (NHL-BFM) multicenter trials. Haematologica 2010, 95, 253–259. [Google Scholar] [CrossRef] [Green Version]
  53. Perkins, S.L.; Gross, T.G. Pediatric indolent lymphoma—Would less be better? Pediatr. Blood Cancer 2011, 57, 189–190. [Google Scholar] [CrossRef] [PubMed]
  54. Tan, D.; Horning, S.J. Follicular Lymphoma: Clinical Features and Treatment. Hematol. Oncol. Clin. N. Am. 2008, 22, 863–882. [Google Scholar] [CrossRef] [PubMed]
  55. Kaseb, H.; Mukkamalla, S.K.R.; Rajasurya, V. Anaplastic Large Cell Lymphoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  56. Turner, S.D.; Lamant, L.; Kenner, L.; Brugières, L. Anaplastic large cell lymphoma in paediatric and young adult patients. Br. J. Haematol. 2016, 173, 560–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Benharroch, D.; Meguerian-Bedoyan, Z.; Lamant, L.; Amin, C.; Brugières, L.; Terrier-Lacombe, M.-J.; Haralambieva, E.; Pulford, K.; Pileri, S.; Morris, S.W.; et al. ALK-Positive Lymphoma: A Single Disease with a Broad Spectrum of Morphology. Blood 1998, 91, 2076–2084. [Google Scholar] [CrossRef]
  58. Le Deley, M.-C.; Reiter, A.; Williams, D.; Delsol, G.; Oschlies, I.; McCarthy, K.; Zimmermann, M.; Brugières, L.; European Intergroup for Childhood Non-Hodgkin Lymphoma. Prognostic factors in childhood anaplastic large cell lymphoma: Results of a large European intergroup study. Blood 2008, 111, 1560–1566. [Google Scholar] [CrossRef] [Green Version]
  59. Mellgren, K.; Hedegaard, C.J.; Schmiegelow, K.; Müller, K. Plasma Cytokine Profiles at Diagnosis in Pediatric Patients with Non-Hodgkin Lymphoma. J. Pediatr. Hematol. Oncol. 2012, 34, 271–275. [Google Scholar] [CrossRef] [Green Version]
  60. Boi, M.; Zucca, E.; Inghirami, G.; Bertoni, F. Advances in understanding the pathogenesis of systemic anaplastic large cell lymphomas. Br. J. Haematol. 2015, 168, 771–783. [Google Scholar] [CrossRef]
  61. Mussolin, L.; Pillon, M.; d’Amore, E.S.; Santoro, N.; Lombardi, A.; Fagioli, F.; Zanesco, L.; Rosolen, A. Prevalence and clinical implications of bone marrow involvement in pediatric anaplastic large cell lymphoma. Leukemia 2005, 19, 1643–1647. [Google Scholar] [CrossRef] [Green Version]
  62. National Cancer Institute Childhood Non-Hodgkin Lymphoma Treatment (PDQ®)—Health Professional Version. Available online: https://www.cancer.gov/types/lymphoma/hp/child-nhl-treatment-pdq#cit/section_6.21 (accessed on 18 December 2021).
  63. Alexander, S.a.F.; Adolfo, A. Pathologic Features: Diffuse Large B-Cell Lymphomas. In Nathan and Oski’s Hematology and Oncology of Infancy and Childhood, 8th ed.; Stuart, H., Orkin, D.E.F., Thomas, L.A., Samuel, E., Lux, D.G., IV, Nathan, D.G., Eds.; Elsevier Health Sciences: Amsterdam, The Netherlands, 2015; p. 1660. [Google Scholar]
  64. El-Mallawany, N.K.; Cairo, M.S. Advances in the diagnosis and treatment of childhood and adolescent B-cell non-Hodgkin lymphoma. Clin. Adv. Hematol. Oncol. 2015, 13, 113–123. [Google Scholar]
  65. Minard-Colin, V.; Brugières, L.; Reiter, A.; Cairo, M.S.; Gross, T.G.; Woessmann, W.; Burkhardt, B.; Sandlund, J.T.; Williams, D.; Pillon, M.; et al. Non-Hodgkin Lymphoma in Children and Adolescents: Progress Through Effective Collaboration, Current Knowledge, and Challenges Ahead. J. Clin. Oncol. 2015, 33, 2963–2974. [Google Scholar] [CrossRef]
  66. Reiter, A.; Schrappe, M.; Tiemann, M.; Parwaresch, R.; Zimmermann, M.; Yakisan, E.; Dopfer, R.; Bucsky, P.; Mann, G.; Gadner, H. Successful treatment strategy for Ki-1 anaplastic large-cell lymphoma of childhood: A prospective analysis of 62 patients enrolled in three consecutive Berlin-Frankfurt-Munster group studies. J. Clin. Oncol. 1994, 12, 899–908. [Google Scholar] [CrossRef] [PubMed]
  67. Alexander, S.; Kraveka, J.M.; Weitzman, S.; Lowe, E.; Smith, L.; Lynch, J.C.; Chang, M.; Kinney, M.C.; Perkins, S.L.; Laver, J.; et al. Advanced stage anaplastic large cell lymphoma in children and adolescents: Results of ANHL0131, a randomized phase III trial of APO versus a modified regimen with vinblastine: A report from the children’s oncology group. Pediatr. Blood Cancer 2014, 61, 2236–2242. [Google Scholar] [CrossRef] [Green Version]
  68. Seidemann, K.; Tiemann, M.; Schrappe, M.; Yakisan, E.; Simonitsch, I.; Janka-Schaub, G.; Dörffel, W.; Zimmermann, M.; Mann, G.; Gadner, H.; et al. Short-pulse B–non-Hodgkin lymphoma–type chemotherapy is efficacious treatment for pediatric anaplastic large cell lymphoma: A report of the Berlin-Frankfurt-Münster Group Trial NHL-BFM 90. Blood 2001, 97, 3699–3706. [Google Scholar] [CrossRef]
  69. Hayashi, R.J.; Winter, S.S.; Dunsmore, K.P.; Devidas, M.; Wood, B.; Hermiston, M.L.; Teachey, D.T.; Perkins, S.L.; Miles, R.R.; Raetz, E.A.; et al. Children’s Oncology Group (COG) AALL0434: Successful Disease Control without Cranial Radiation in Newly Diagnosed T Lymphoblastic Lymphoma (T-LL). Blood 2018, 132 (Suppl. 1), 1000. [Google Scholar] [CrossRef]
  70. Chang, J.E.; Medlin, S.C.; Kahl, B.S.; Longo, W.L.; Williams, E.C.; Lionberger, J.; Kim, K.; Kim, J.; Esterberg, E.; Juckett, M.B. Augmented and standard Berlin-Frankfurt-Munster chemotherapy for treatment of adult acute lymphoblastic leukemia. Leuk. Lymphoma 2008, 49, 2298–2307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Rytting, M.E.; Thomas, D.A.; O’Brien, S.M.; Ravandi-Kashani, F.; Jabbour, E.J.; Franklin, A.R.; Kadia, T.M.; Pemmaraju, N.; Daver, N.G.; Ferrajoli, A.; et al. Augmented Berlin-Frankfurt-Münster therapy in adolescents and young adults (AYAs) with acute lymphoblastic leukemia (ALL). Cancer 2014, 120, 3660–3668. [Google Scholar] [CrossRef] [PubMed]
  72. Dunsmore, K.P.; Winter, S.S.; Devidas, M.; Wood, B.L.; Esiashvili, N.; Chen, Z.; Eisenberg, N.; Briegel, N.; Hayashi, R.J.; Gastier-Foster, J.M.; et al. Children’s Oncology Group AALL0434: A Phase III Randomized Clinical Trial Testing Nelarabine in Newly Diagnosed T-Cell Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2020, 38, 3282–3293. [Google Scholar] [CrossRef]
  73. Swerdlow, S.; Campo, E.; Harris, N.; Jaffe, E.; Pileri, S.; Stein, H.; Thiele, J.; Arber, D.; Hasserjian, R.; Le Beau, M. WHO Classification of Tumors of Hematopoietic and Lymphoid Tissues, Revised, 4th ed.; IARC: Lyon, France, 2017. [Google Scholar]
  74. Swerdlow, S.H.; Campo, E.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.; Thiele, J.; Vardiman, J.W. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues; International Agency for Research on Cancer: Lyon, France, 2008; Volume 2. [Google Scholar]
  75. Chisholm, K.M.; Mohlman, J.; Liew, M.; Termuhlen, A.; Cairo, M.S.; Gross, T.G.; Perkins, S.L.; Miles, R.R. IRF4 translocation status in pediatric follicular and diffuse large B-cell lymphoma patients enrolled in Children’s Oncology Group trials. Pediatr. Blood Cancer 2019, 66, e27770. [Google Scholar] [CrossRef]
  76. Patte, C.; Auperin, A.; Gerrard, M.; Michon, J.; Pinkerton, R.; Sposto, R.; Weston, C.; Raphael, M.; Perkins, S.L.; McCarthy, K.; et al. Results of the randomized international FAB/LMB96 trial for intermediate risk B-cell non-Hodgkin lymphoma in children and adolescents: It is possible to reduce treatment for the early responding patients. Blood 2006, 109, 2773–2780. [Google Scholar] [CrossRef] [Green Version]
  77. Minard-Colin, V.; Aupérin, A.; Pillon, M.; Burke, G.A.A.; Barkauskas, D.A.; Wheatley, K.; Delgado, R.F.; Alexander, S.; Uyttebroeck, A.; Bollard, C.M.; et al. Rituximab for High-Risk, Mature B-Cell Non-Hodgkin’s Lymphoma in Children. N. Engl. J. Med. 2020, 382, 2207–2219. [Google Scholar] [CrossRef]
  78. Giulino-Roth, L.; O’Donohue, T.; Chen, Z.; Bartlett, N.L.; LaCasce, A.; Martin-Doyle, W.; Barth, M.J.; Davies, K.; Blum, K.A.; Christian, B.; et al. Outcomes of adults and children with primary mediastinal B-cell lymphoma treated with dose-adjusted EPOCH-R. Br. J. Haematol. 2017, 179, 739–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Burke, G.A.A.; Minard-Colin, V.; Aupérin, A.; Alexander, S.; Pillon, M.; Delgado, R.; Zsíros, J.; Uyttebroeck, A.; Dartigues, P.; Miles, R.R.; et al. Dose-Adjusted Etoposide, Doxorubicin, and Cyclophosphamide with Vincristine and Prednisone Plus Rituximab Therapy in Children and Adolescents with Primary Mediastinal B-Cell Lymphoma: A Multicenter Phase II Trial. J. Clin. Oncol. 2021, 39, 3716–3724. [Google Scholar] [CrossRef] [PubMed]
  80. Seidemann, K.; Tiemann, M.; Lauterbach, I.; Mann, G.; Simonitsch, I.; Stankewitz, K.; Schrappe, M.; Zimmermann, M.; Niemeyer, C.; Parwaresch, R.; et al. Primary Mediastinal Large B-Cell Lymphoma with Sclerosis in Pediatric and Adolescent Patients: Treatment and Results From Three Therapeutic Studies of the Berlin-Frankfurt-Münster Group. J. Clin. Oncol. 2003, 21, 1782–1789. [Google Scholar] [CrossRef]
  81. Prokoph, N.; Larose, H.; Lim, M.S.; Burke, G.A.A.; Turner, S.D. Treatment Options for Paediatric Anaplastic Large Cell Lymphoma (ALCL): Current Standard and beyond. Cancers 2018, 10, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Brugières, L.; Deley, M.C.L.; Pacquement, H.; Meguerian-Bedoyan, Z.; Terrier-Lacombe, M.J.; Robert, A.; Pondarré, C.; Leverger, G.; Devalck, C.; Rodary, C.; et al. CD30+ Anaplastic Large-Cell Lymphoma in Children: Analysis of 82 Patients Enrolled in Two Consecutive Studies of the French Society of Pediatric Oncology. Blood 1998, 92, 3591–3598. [Google Scholar]
  83. Kraveka, J.M.; Weitzman, S.S.; Smith, L.; Lynch, J.C.; Chang, M.; Kinney, M.; Perkins, S.L.; Laver, J.H.; Gross, T.G.; Weinstein, H.J.; et al. Advanced-stage anaplastic large-cell lymphoma in children and adolescents: Results of ANHL0131, a randomized phase III trial with standard APO (doxorubicin, prednisone, vincristine) versus consolidation with a regimen including vinblastine: A report from the Children’s Oncology Group. J. Clin. Oncol. 2010, 28 (Suppl. 15), 9511. [Google Scholar] [CrossRef]
  84. Reiter, A.; Schrappe, M.; Ludwig, W.D.; Tiemann, M.; Parwaresch, R.; Zimmermann, M.; Schirg, E.; Henze, G.; Schellong, G.; Gadner, H.; et al. Intensive ALL-type therapy without local radiotherapy provides a 90% event-free survival for children with T-cell lymphoblastic lymphoma: A BFM group report. Blood 2000, 95, 416–421. [Google Scholar]
  85. Teachey, D.T.; O’Connor, D. How I treat newly diagnosed T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma in children. Blood 2020, 135, 159–166. [Google Scholar] [CrossRef]
  86. Kumar, R.; Galardy, P.J.; Dogan, A.; Rodriguez, V.; Khan, S.P. Rituximab in combination with multiagent chemotherapy for pediatric follicular lymphoma. Pediatr. Blood Cancer 2011, 57, 317–320. [Google Scholar] [CrossRef]
  87. Moleti, M.L.; Testi, A.M.; Foà, R. Treatment of relapsed/refractory paediatric aggressive B-cell non-Hodgkin lymphoma. Br. J. Haematol. 2020, 189, 826–843. [Google Scholar] [CrossRef]
  88. Jourdain, A.; Auperin, A.; Minard-Colin, V.; Aladjidi, N.; Zsiros, J.; Coze, C.; Gandemer, V.; Bertrand, Y.; Leverger, G.; Bergeron, C.; et al. Outcome of and prognostic factors for relapse in children and adolescents with mature B-cell lymphoma and leukemia treated in three consecutive prospective “Lymphomes Malins B” protocols. A Société Française des Cancers de l’Enfant study. Haematologica 2015, 100, 810–817. [Google Scholar] [CrossRef] [PubMed]
  89. Burkhardt, B.; Taj, M.; Garnier, N.; Minard-Colin, V.; Hazar, V.; Mellgren, K.; Osumi, T.; Fedorova, A.; Myakova, N.; Verdu-Amoros, J.; et al. Treatment and Outcome Analysis of 639 Relapsed Non-Hodgkin Lymphomas in Children and Adolescents and Resulting Treatment Recommendations. Cancers 2021, 13, 2075. [Google Scholar] [CrossRef] [PubMed]
  90. Rigaud, C.; Auperin, A.; Jourdain, A.; Haouy, S.; Couec, M.-L.; Aladjidi, N.; Gandemer, V.; Lambliotte, A.; Plat, G.; Landman-Parker, J.; et al. Outcome of relapse in children and adolescents with B-cell non-Hodgkin lymphoma and mature acute leukemia: A report from the French LMB study. Pediatr. Blood Cancer 2019, 66, e27873. [Google Scholar] [CrossRef]
  91. Osumi, T.; Mori, T.; Fujita, N.; Saito, A.M.; Nakazawa, A.; Tsurusawa, M.; Kobayashi, R. Relapsed/refractory pediatric B-cell non-Hodgkin lymphoma treated with rituximab combination therapy: A report from the Japanese Pediatric Leukemia/Lymphoma Study Group. Pediatr. Blood Cancer 2016, 63, 17949. [Google Scholar] [CrossRef] [PubMed]
  92. Griffin, T.C.; Weitzman, S.; Weinstein, H.; Chang, M.; Cairo, M.; Hutchison, R.; Shiramizu, B.; Wiley, J.; Woods, D.; Barnich, M.; et al. A study of rituximab and ifosfamide, carboplatin, and etoposide chemotherapy in children with recurrent/refractory B-cell (CD20+) non-Hodgkin lymphoma and mature B-cell acute lymphoblastic leukemia: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2009, 52, 177–181. [Google Scholar] [CrossRef] [Green Version]
  93. Gross, T.G.; Hale, G.A.; He, W.; Camitta, B.M.; Sanders, J.E.; Cairo, M.S.; Hayashi, R.J.; Termuhlen, A.M.; Zhang, M.J.; Davies, S.M.; et al. Hematopoietic stem cell transplantation for refractory or recurrent non-Hodgkin lymphoma in children and adolescents. Biol. Blood Marrow Transplant. 2010, 16, 223–230. [Google Scholar] [CrossRef] [Green Version]
  94. Giulino-Roth, L.; Ricafort, R.; Kernan, N.A.; Small, T.N.; Trippett, T.M.; Steinherz, P.G.; Prockop, S.E.; Scaradavou, A.; Chiu, M.; O’Reilly, R.J.; et al. Ten-year follow-up of pediatric patients with non-Hodgkin lymphoma treated with allogeneic or autologous stem cell transplantation. Pediatr. Blood Cancer 2013, 60, 2018–2024. [Google Scholar] [CrossRef]
  95. Strullu, M.; Thomas, C.; Le Deley, M.C.; Chevance, A.; Kanold, J.; Bertrand, Y.; Jubert, C.; Dalle, J.H.; Paillard, C.; Baruchel, A.; et al. Hematopoietic stem cell transplantation in relapsed ALK+ anaplastic large cell lymphoma in children and adolescents: A study on behalf of the SFCE and SFGM-TC. Bone Marrow Transplant. 2015, 50, 795–801. [Google Scholar] [CrossRef]
  96. Brugières, L.; Quartier, P.; Le Deley, M.C.; Pacquement, H.; Perel, Y.; Bergeron, C.; Schmitt, C.; Landmann, J.; Patte, C.; Terrier-Lacombe, M.J.; et al. Relapses of childhood anaplastic large-cell lymphoma: Treatment results in a series of 41 children—A report from the French Society of Pediatric Oncology. Ann. Oncol. 2000, 11, 53–58. [Google Scholar] [CrossRef]
  97. Naik, S.; Martinez, C.A.; Omer, B.; Sasa, G.; Yassine, K.; Allen, C.E.; Kamdar, K.; Orth, R.; Wu, M.; Leung, K.; et al. Allogeneic hematopoietic stem cell transplant for relapsed and refractory non-Hodgkin lymphoma in pediatric patients. Blood Adv. 2019, 3, 2689–2695. [Google Scholar] [CrossRef]
  98. Cairo, M.S.; Beishuizen, A. Childhood, adolescent and young adult non-Hodgkin lymphoma: Current perspectives. Br. J. Haematol. 2019, 185, 1021–1042. [Google Scholar] [CrossRef] [PubMed]
  99. Butcher, B.W.; Collins, R.H. The graft-versus-lymphoma effect: Clinical review and future opportunities. Bone Marrow Transplant. 2005, 36, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Maloney, D.G. Graft-vs.-lymphoma effect in various histologies of non-Hodgkin’s lymphoma. Leuk. Lymphoma 2003, 44 (Suppl. 3), S99–S105. [Google Scholar] [CrossRef] [PubMed]
  101. Bishop, M.R.; Dean, R.M.; Steinberg, S.M.; Odom, J.; Pavletic, S.Z.; Chow, C.; Pittaluga, S.; Sportes, C.; Hardy, N.M.; Gea-Banacloche, J.; et al. Clinical evidence of a graft-versus-lymphoma effect against relapsed diffuse large B-cell lymphoma after allogeneic hematopoietic stem-cell transplantation. Ann. Oncol. 2008, 19, 1935–1940. [Google Scholar] [CrossRef]
  102. Jeon, Y.-W.; Yoon, S.; Min, G.J.; Park, S.-S.; Park, S.; Yoon, J.-H.; Lee, S.-E.; Cho, B.-S.; Eom, K.-S.; Kim, Y.-J.; et al. Risk factors predicting graft-versus-host disease and relapse-free survival after allogeneic hematopoietic stem cell transplantation in relapsed or refractory non-Hodgkin’s lymphoma. Ann. Hematol. 2019, 98, 1743–1753. [Google Scholar] [CrossRef] [Green Version]
  103. Storb, R.; Gyurkocza, B.; Storer, B.E.; Sorror, M.L.; Blume, K.; Niederwieser, D.; Chauncey, T.R.; Pulsipher, M.A.; Petersen, F.B.; Sahebi, F.; et al. Graft-versus-host disease and graft-versus-tumor effects after allogeneic hematopoietic cell transplantation. J. Clin. Oncol. 2013, 31, 1530–1538. [Google Scholar] [CrossRef]
  104. Harker-Murray, P.D.; Pommert, L.; Barth, M.J. Novel Therapies Potentially Available for Pediatric B-Cell Non-Hodgkin Lymphoma. J. Natl. Compr. Cancer Netw. 2020, 18, 1125–1134. [Google Scholar] [CrossRef]
  105. Genentech Inc. RITUXAN® (Rituxan) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/103705s5467lbl.pdf (accessed on 30 December 2021).
  106. Minard, V.; Gross, T. Results of the Randomized Intergroup Trial Inter-B-NHL Ritux 2010 for Children and Adolescents with High-Risk B-Cell Non-Hodgkin lymphoma (B-NHL) and Mature Acute Leukemia (B-AL): Evaluation of Rituximab (R) Efficacy in Addition to Standard LMB Chemotherapy (CT) Regimen; Gustave Roussy, Cancer Campus, Grand Paris, Children’s Oncology Group; American Society of Clinical Oncology: Alexandria, VA, USA, 2016. [Google Scholar]
  107. Barth, M.J.; Goldman, S.; Smith, L.; Perkins, S.L.; Shiramizu, B.; Gross, T.G.; Harrison, L.; Sanger, W.; Geyer, M.B.; Giulino, L.B. Rituximab Pharmacokinetics in Children and Adolescents with de Novo Intermediate and Advanced mature B-Cell Lymphoma/Leukemia: A Children’s Oncology Group (COG) Report; American Society of Clinical Oncology: Alexandria, VA, USA, 2013. [Google Scholar]
  108. Seattle Genetics Inc. ADCETRIS® (Brentuximab Vedotin) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125388s097lbl.pdf (accessed on 30 December 2021).
  109. Lowe, E.J.; Reilly, A.F.; Lim, M.S.; Gross, T.G.; Saguilig, L.; Barkauskas, D.A.; Wu, R.; Alexander, S.; Bollard, C.M. Brentuximab Vedotin in Combination with Chemotherapy for Pediatric Patients with ALK+ ALCL: Results of COG Trial ANHL12P1. Blood 2021, 137, 3595–3603. [Google Scholar] [CrossRef]
  110. Shustov, A. Ceritinib with Brentuximab Vedotin in Treating Patients with ALK-Positive Anaplastic Large Cell Lymphoma; Last updated October 2019; National Cancer Institute (NCI): Bethesda, MD, USA, 2019.
  111. Zinzani, P.L.; Santoro, A.; Gritti, G.; Brice, P.; Barr, P.M.; Kuruvilla, J.; Cunningham, D.; Kline, J.; Johnson, N.A.; Mehta-Shah, N.; et al. Nivolumab Combined with Brentuximab Vedotin for Relapsed/Refractory Primary Mediastinal Large B-Cell Lymphoma: Efficacy and Safety From the Phase II CheckMate 436 Study. J. Clin. Oncol. 2019, 37, 3081–3089. [Google Scholar] [CrossRef]
  112. Brugieres, L. Nivolumab for Pediatric and Adult Relapsing/Refractory ALK+, for Evaluation of Response in Patients with Progressive Disease (Cohort 1) or as Consolidative Immunotherapy in Patients in Complete Remission after Relapse (Cohort 2); Last updated July 2021; Gustave Roussy, Cancer Campus: Paris, France, 2021. [Google Scholar]
  113. Amgen Inc. BLINCYTO® (Blinatumomab) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125557s013lbl.pdf (accessed on 30 December 2021).
  114. Bargou, R. Safety Study of the Bispecific T-Cell Engager Blinatumomab (MT103) in Patients with Relapsed NHL; Last updated January 2015; Amgen Research (Munich) GmbH: Munich, Germany, 2015. [Google Scholar]
  115. Amgen Research (Munich) Gmb. Clinical Study with Blinatumomab in Patients with Relapsed/Refractory Diffuse Large B-Cell Lymphoma (DLBCL); Last Updated January 2017; Amgen Research (Munich) GmbH: Munich, Germany, 2017. [Google Scholar]
  116. Breese, E. Pembro + Blina Combination in Pediatric and Young Adult Patients with Relapsed/Refractory Acute Leukemia or Lymphoma; Last updated April 2021; Children’s Hospital Medical Center: Cincinnati, OH, USA, 2021. [Google Scholar]
  117. Inaba, H. Total Therapy XVII for Newly Diagnosed Patients with Acute Lymphoblastic Leukemia and Lymphoma; Last updated August 2021; St. Jude Children’s Research Hospital: Memphis, TN, USA, 2021. [Google Scholar]
  118. Wyeth Pharmaceuticals Inc.; Subsidiary of Pfizer Inc. BESPONSA™ (Inotuzumab Ozogamicin) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761040s000lbl.pdf (accessed on 30 December 2021).
  119. O’Brien, M.M. Inotuzumab Ozogamicin in Treating Younger Patients with B-Lymphoblastic Lymphoma or Relapsed or Refractory CD22 Positive B Acute Lymphoblastic Leukemia; Last updated December 2016; Children’s Oncology Group: Seattle, WA, USA, 2016. [Google Scholar]
  120. Pennesi, E.; Michels, N.; Brivio, E.; van der Velden, V.H.J.; Jiang, Y.; Thano, A.; Ammerlaan, A.J.C.; Boer, J.M.; Beverloo, H.B.; Sleight, B.; et al. Inotuzumab ozogamicin as single agent in pediatric patients with relapsed and refractory acute lymphoblastic leukemia: Results from a phase II trial. Leukemia 2022, 36, 1516–1524. [Google Scholar] [CrossRef]
  121. GlaxoSmithKline, ARRANON® (Nelarabine) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/021877s009lbl.pdf (accessed on 30 December 2021).
  122. Winter, S.S. Combination Chemotherapy in Treating Young Patients with Newly Diagnosed T-Cell Acute Lymphoblastic Leukemia or T-Cell Lymphoblastic Lymphoma; Last updated September 2021; National Cancer Institute (NCI): Bethesda, MD, USA, 2021. [Google Scholar]
  123. Novartis Pharmaceuticals Corporation. KYMRIAH® (Tisagenlecleucel) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.fda.gov/media/107296/download (accessed on 30 December 2021).
  124. National Library of Medicine (U.S.). (August 1, 2018) Phase II Open Label Trial to Determine Safety & Efficacy of Tisagenlecleucel in Pediatric Non-Hodgkin Lymphoma Patients. Identifier: NCT03610724. Last updated May 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03610724 (accessed on 28 December 2021).
  125. Kite Pharma Inc. YESCARTA® (Axicabtagene Ciloleucel) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.fda.gov/media/108377/download (accessed on 30 December 2021).
  126. National Library of Medicine (U.S.). (December 9, 2015) Study Evaluating Brexucabtagene Autoleucel (KTE-X19) in Pediatric and Adolescent Participants with Relapsed/Refractory B-precursor Acute Lymphoblastic Leukemia or Relapsed/Refractory B-Cell Non-Hodgkin Lymphoma. Identifier: NCT02625480. Last updated May 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT02625480 (accessed on 28 December 2021).
  127. Shah, N.N. Anti-CD22 Chimeric Receptor T Cells in Pediatric and Young Adults with Recurrent or Refractory CD22-Expressing B Cell Malignancies; Last updated December 2021; National Cancer Institute (NCI): Bethesda, MD, USA, 2021.
  128. Schultz, L. CD22-CAR T Cells in Children and Young Adults with B Cell Malignancies; Last updated May 2021; Stanford University: Stanford, CA, USA, 2021. [Google Scholar]
  129. Shah, N.N. CD19/CD22 Chimeric Antigen Receptor (CAR) T Cells in Children and Young Adults with Recurrent or Refractory CD19/CD22-expressing B Cell Malignancies; Last updated December 2021; National Cancer Institute: Bethesda, MD, USA, 2021.
  130. Heslop, H.E. LMP-Specific T-Cells for Patients with Relapsed EBV-Positive Lymphoma; Last updated July 2020; Baylor College of Medicine, The Methodist Hospital Research Institute, Center for Cell and Gene Therapy, Baylor College of Medicine: Houston, TX, USA, 2020. [Google Scholar]
  131. Hu, Y. Cord Blood Derived Anti-CD19 CAR-Engineered NK Cells for B Lymphoid Malignancies; Last updated April 2021; Wuhan Union Hospital: Wuhan, China, 2021. [Google Scholar]
  132. Nieto, Y.L. Umbilical Cord Blood NK Cells, Rituximab, High-Dose Chemotherapy, and Stem Cell Transplant in Treating Patients with Recurrent or Refractory B-Cell Non-Hodgkin’s Lymphoma; Last updated January 2017; National Cancer Institute (NCI): Bethesda, MD, USA, 2021.
  133. Gozdzik, J.; Rewucka, K.; Krasowska-Kwiecien, A.; Pieczonka, A.; Debski, R.; Zaucha-Prazmo, A.; Drabko, K.; Krukowska-Jaros, J.; Wozniak, M.; Kowalczyk, J. Adoptive therapy with donor lymphocyte infusion after allogenic hematopoietic SCT in pediatric patients. Bone Marrow Transplant. 2015, 50, 51–55. [Google Scholar] [CrossRef] [Green Version]
  134. Merck Sharp & Dohme Corp. A Subsidiary of Merck & Co. Inc. KEYTRUDA® (Pembrolizumab) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.fda.gov/media/108377/download (accessed on 30 December 2021).
  135. Geoerger, B.; Kang, H.J.; Yalon-Oren, M.; Marshall, L.V.; Vezina, C.; Pappo, A.; Laetsch, T.W.; Petrilli, A.S.; Ebinger, M.; Toporski, J.; et al. Pembrolizumab in paediatric patients with advanced melanoma or a PD-L1-positive, advanced, relapsed, or refractory solid tumour or lymphoma (KEYNOTE-051): Interim analysis of an open-label, single-arm, phase 1-2 trial. Lancet Oncol. 2020, 21, 121–133. [Google Scholar] [CrossRef]
  136. Bristol-Myers Squibb Company. OPDIVO (Nivolumab) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/125554s090lbl.pdf (accessed on 30 December 2021).
  137. Davis, K.L.; Fox, E.; Merchant, M.S.; Reid, J.M.; Kudgus, R.A.; Liu, X.; Minard, C.G.; Voss, S.; Berg, S.L.; Weigel, B.J.; et al. Nivolumab in children and young adults with relapsed or refractory solid tumours or lymphoma (ADVL1412): A multicentre, open-label, single-arm, phase 1–2 trial. Lancet Oncol. 2020, 21, 541–550. [Google Scholar] [CrossRef]
  138. Bristol-Myers, S. A Study of Nivolumab Plus Brentuximab Vedotin in Patients between 5 and 30 Years Old, with Hodgkin’s Lymphoma (cHL), Relapsed or Refractory from First Line Treatment. CheckMate 744: CHECKpoint Pathway and Nivolumab Clinical Trial Evaluation); Last updated June 2020; Seagen Inc.: Washington, DC, USA, 2020. [Google Scholar]
  139. Genentech Inc. TECENTRIQ® (Atezolizumab) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761034s042lbl.pdf (accessed on 30 December 2021).
  140. National Library of Medicine (U.S.). (September 4, 2015) A Study to Evaluate the Safety, Tolerability, Pharmacokinetics, Immunogenicity, and Preliminary Efficacy of Atezolizumab (Anti-Programmed Death-Ligand 1 PD-L1 Antibody) in Pediatric and Young Adult Participants with Solid Tumors. Identifier: NCT02541604. Last updated February 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT02541604 (accessed on 29 December 2021).
  141. Pfizer Inc. XALKORI® (Crizotinib) [Package Insert]. U.S. Food and Drug Administration Website. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/202570s030lbl.pdf (accessed on 1 December 2021).
  142. Mossé, Y.P.; Lim, M.S.; Voss, S.D.; Wilner, K.; Ruffner, K.; Laliberte, J.; Rolland, D.; Balis, F.M.; Maris, J.M.; Weigel, B.J. Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: A Children’s Oncology Group phase 1 consortium study. Lancet Oncol. 2013, 14, 472–480. [Google Scholar] [CrossRef] [Green Version]
  143. Nie, M.; Wang, Y.; Bi, X.-W.; Xia, Y.; Sun, P.; Liu, P.-P.; Li, Z.-M.; Jiang, W.-Q. Effect of rituximab on adult Burkitt’s lymphoma: A systematic review and meta-analysis. Ann. Hematol. 2016, 95, 19–26. [Google Scholar] [CrossRef] [PubMed]
  144. Goldman, S.; Smith, L.; Anderson, J.R.; Perkins, S.; Harrison, L.; Geyer, M.B.; Gross, T.G.; Weinstein, H.; Bergeron, S.; Shiramizu, B.; et al. Rituximab and FAB/LMB 96 chemotherapy in children with Stage III/IV B-cell non-Hodgkin lymphoma: A Children’s Oncology Group report. Leukemia 2013, 27, 1174–1177. [Google Scholar] [CrossRef] [Green Version]
  145. Frazer, J.K.; Li, K.J.; Galardy, P.J.; Perkins, S.L.; Auperin, A.; Anderson, J.R.; Pinkerton, R.; Buxton, A.; Gross, T.G.; Michon, J.; et al. Excellent outcomes in children and adolescents with CNS(+) Burkitt lymphoma or other mature B-NHL using only intrathecal and systemic chemoimmunotherapy: Results from FAB/LMB96 and COG ANHL01P1. Br. J. Haematol. 2019, 185, 374–377. [Google Scholar] [CrossRef] [Green Version]
  146. Goldman, S.; Smith, L.; Galardy, P.; Perkins, S.L.; Frazer, J.K.; Sanger, W.; Anderson, J.R.; Gross, T.G.; Weinstein, H.; Harrison, L.; et al. Rituximab with chemotherapy in children and adolescents with central nervous system and/or bone marrow-positive Burkitt lymphoma/leukaemia: A Children’s Oncology Group Report. Br. J. Haematol. 2014, 167, 394–401. [Google Scholar] [CrossRef]
  147. Leppä, S.; Jørgensen, J.; Tierens, A.; Meriranta, L.; Østlie, I.; de Nully Brown, P.; Fagerli, U.-M.; Larsen, T.S.; Mannisto, S.; Munksgaard, L.; et al. Patients with high-risk DLBCL benefit from dose-dense immunochemotherapy combined with early systemic CNS prophylaxis. Blood Adv. 2020, 4, 1906–1915. [Google Scholar] [CrossRef]
  148. Goldman, S.; Barth, M.; Shiramizu, B.; Shi, Q.; Hochberg, J.; Klejmont, L.; Harrison, L.; Basso, J.; Chu, Y.; Islam, H.; et al. A dose substitution of anthracycline intensity with dose-dense rituximab in children and adolescents with good-risk mature B-cell lymphoma. Leukemia 2021, 35, 2994–2997. [Google Scholar] [CrossRef]
  149. U.S. Food and Drug Administration. FDA Approves Rituximab Plus Chemotherapy for Pediatric Cancer Indications. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-rituximab-plus-chemotherapy-pediatric-cancer-indications (accessed on 30 December 2021).
  150. Berger, G.K.; McBride, A.; Lawson, S.; Royball, K.; Yun, S.; Gee, K.; Bin Riaz, I.; Saleh, A.A.; Puvvada, S.; Anwer, F. Brentuximab vedotin for treatment of non-Hodgkin lymphomas: A systematic review. Crit. Rev. Oncol. Hematol. 2017, 109, 42–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Locatelli, F.; Mauz-Koerholz, C.; Neville, K.; Llort, A.; Beishuizen, A.; Daw, S.; Pillon, M.; Aladjidi, N.; Klingebiel, T.; Landman-Parker, J.; et al. Brentuximab vedotin for paediatric relapsed or refractory Hodgkin’s lymphoma and anaplastic large-cell lymphoma: A multicentre, open-label, phase 1/2 study. Lancet Haematol. 2018, 5, e450–e461. [Google Scholar] [CrossRef]
  152. Locatelli, F.; Zugmaier, G.; Rizzari, C.; Morris, J.D.; Gruhn, B.; Klingebiel, T.; Parasole, R.; Linderkamp, C.; Flotho, C.; Petit, A.; et al. Effect of Blinatumomab vs Chemotherapy on Event-Free Survival Among Children with High-risk First-Relapse B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA 2021, 325, 843–854. [Google Scholar] [CrossRef] [PubMed]
  153. Inaba, H.; Pui, C.H. Immunotherapy in pediatric acute lymphoblastic leukemia. Cancer Metastasis Rev. 2019, 38, 595–610. [Google Scholar] [CrossRef] [PubMed]
  154. Dufner, V.; Sayehli, C.M.; Chatterjee, M.; Hummel, H.D.; Gelbrich, G.; Bargou, R.C.; Goebeler, M.-E. Long-term outcome of patients with relapsed/refractory B-cell non-Hodgkin lymphoma treated with blinatumomab. Blood Adv. 2019, 3, 2491–2498. [Google Scholar] [CrossRef] [Green Version]
  155. Goebeler, M.E.; Knop, S.; Viardot, A.; Kufer, P.; Topp, M.S.; Einsele, H.; Noppeney, R.; Hess, G.; Kallert, S.; Mackensen, A.; et al. Bispecific T-Cell Engager (BiTE) Antibody Construct Blinatumomab for the Treatment of Patients with Relapsed/Refractory Non-Hodgkin Lymphoma: Final Results From a Phase I Study. J. Clin. Oncol. 2016, 34, 1104–1111. [Google Scholar] [CrossRef] [PubMed]
  156. Viardot, A.; Goebeler, M.E.; Hess, G.; Neumann, S.; Pfreundschuh, M.; Adrian, N.; Zettl, F.; Libicher, M.; Sayehli, C.; Stieglmaier, J.; et al. Phase 2 study of the bispecific T-cell engager (BiTE) antibody blinatumomab in relapsed/refractory diffuse large B-cell lymphoma. Blood 2016, 127, 1410–1416. [Google Scholar] [CrossRef]
  157. Coyle, L.; Morley, N.J.; Rambaldi, A.; Mason, K.D.; Verhoef, G.; Furness, C.L.; Zhang, A.; Jung, A.S.; Cohan, D.; Franklin, J.L. Open-Label, phase 2 study of blinatumomab as second salvage therapy in adults with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma. Leuk. Lymphoma 2020, 61, 2103–2112. [Google Scholar] [CrossRef]
  158. Bhojwani, D.; Sposto, R.; Shah, N.N.; Rodriguez, V.; Yuan, C.; Stetler-Stevenson, M.; O’Brien, M.M.; McNeer, J.L.; Quereshi, A.; Cabannes, A.; et al. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Leukemia 2019, 33, 884–892. [Google Scholar] [CrossRef]
  159. Goy, A.; Forero, A.; Wagner-Johnston, N.; Christopher Ehmann, W.; Tsai, M.; Hatake, K.; Ananthakrishnan, R.; Volkert, A.; Vandendries, E.; Ogura, M. A phase 2 study of inotuzumab ozogamicin in patients with indolent B-cell non-Hodgkin lymphoma refractory to rituximab alone, rituximab and chemotherapy, or radioimmunotherapy. Br. J. Haematol. 2016, 174, 571–581. [Google Scholar] [CrossRef]
  160. Kebriaei, P.; Cutler, C.; de Lima, M.; Giralt, S.; Lee, S.J.; Marks, D.; Merchant, A.; Stock, W.; van Besien, K.; Stelljes, M. Management of important adverse events associated with inotuzumab ozogamicin: Expert panel review. Bone Marrow Transplant. 2018, 53, 449–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Salvaris, R.; Ong, J.; Gregory, G.P. Bispecific Antibodies: A Review of Development, Clinical Efficacy and Toxicity in B-Cell Lymphomas. J. Pers. Med. 2021, 11, 355. [Google Scholar] [CrossRef] [PubMed]
  162. Ghosh, N.; Townsend, W.; Dickinson, M.; Topp, M.S.; Tani, M.; Santoro, A.; Crump, M.; Morschhauser, F.; Le Gouill, S.; Mehta, A.; et al. Glofitamab Plus R-CHOP Induces High Response Rates with Minimal Cytokine Release Syndrome (CRS) in Patients (pts) with Relapsed/Refractory (R/R) Non-Hodgkin Lymphoma (NHL) and Previously Untreated (1L) Diffuse Large B-Cell Lymphoma (DLBCL): Preliminary Results from a Dose-Escalation and Safety Run-in Phase Ib Study; American Society of Hematology Annual Meeting and Exposition: Atlanta, GA, USA, 2021. [Google Scholar]
  163. Budde, L.E.; Assouline, S.; Sehn, L.H.; Schuster, S.J.; Yoon, S.-S.; Yoon, D.H.; Matasar, M.J.; Bosch, F.; Kim, W.S.; Nastoupil, L.J.; et al. Single-Agent Mosunetuzumab Shows Durable Complete Responses in Patients with Relapsed or Refractory B-Cell Lymphomas: Phase I Dose-Escalation Study. J. Clin. Oncol. 2022, 40, 481–491. [Google Scholar] [CrossRef]
  164. Kisor, D.F.; Plunkett, W.; Kurtzberg, J.; Mitchell, B.; Hodge, J.P.; Ernst, T.; Keating, M.J.; Gandhi, V. Pharmacokinetics of nelarabine and 9-beta-D-arabinofuranosyl guanine in pediatric and adult patients during a phase I study of nelarabine for the treatment of refractory hematologic malignancies. J. Clin. Oncol. 2000, 18, 995–1003. [Google Scholar] [CrossRef] [PubMed]
  165. Berg, S.L.; Blaney, S.M.; Devidas, M.; Lampkin, T.A.; Murgo, A.; Bernstein, M.; Billett, A.; Kurtzberg, J.; Reaman, G.; Gaynon, P.; et al. Phase II Study of Nelarabine (compound 506U78) in Children and Young Adults with Refractory T-Cell Malignancies: A Report From the Children’s Oncology Group. J. Clin. Oncol. 2005, 23, 3376–3382. [Google Scholar] [CrossRef]
  166. Cohen, M.H.; Johnson, J.R.; Justice, R.; Pazdur, R. FDA Drug Approval Summary: Nelarabine (Arranon®) for the Treatment of T-Cell Lymphoblastic Leukemia/Lymphoma. Oncologist 2008, 13, 709–714. [Google Scholar] [CrossRef]
  167. Hayashi, R.J.; Winter, S.S.; Dunsmore, K.P.; Devidas, M.; Chen, Z.; Wood, B.L.; Hermiston, M.L.; Teachey, D.T.; Perkins, S.L.; Miles, R.R.; et al. Successful Outcomes of Newly Diagnosed T Lymphoblastic Lymphoma: Results From Children’s Oncology Group AALL0434. J. Clin. Oncol. 2020, 38, 3062–3070. [Google Scholar] [CrossRef]
  168. Zwaan, C.M.; Kowalczyk, J.; Schmitt, C.; Bielorai, B.; Russo, M.W.; Woessner, M.; Ranganathan, S.; Leverger, G. Safety and efficacy of nelarabine in children and young adults with relapsed or refractory T-lineage acute lymphoblastic leukaemia or T-lineage lymphoblastic lymphoma: Results of a phase 4 study. Br. J. Haematol. 2017, 179, 284–293. [Google Scholar] [CrossRef]
  169. Kadia, T.M.; Gandhi, V. Nelarabine in the treatment of pediatric and adult patients with T-cell acute lymphoblastic leukemia and lymphoma. Expert Rev. Hematol. 2017, 10, 1–8. [Google Scholar] [CrossRef] [Green Version]
  170. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  171. Larson, R.C.; Maus, M.V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 2021, 21, 145–161. [Google Scholar] [CrossRef] [PubMed]
  172. Yin, Z.; Zhang, Y.; Wang, X. Advances in chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Biomark. Res. 2021, 9, 58. [Google Scholar] [CrossRef] [PubMed]
  173. Locke, F.L.; Miklos, D.B.; Jacobson, C.A.; Perales, M.-A.; Kersten, M.-J.; Oluwole, O.O.; Ghobadi, A.; Rapoport, A.P.; McGuirk, J.; Pagel, J.M.; et al. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N. Engl. J. Med. 2021, 386, 640–654. [Google Scholar] [CrossRef] [PubMed]
  174. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2018, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
  175. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
  176. Shah, B.D.; Ghobadi, A.; Oluwole, O.O.; Logan, A.C.; Boissel, N.; Cassaday, R.D.; Leguay, T.; Bishop, M.R.; Topp, M.S.; Tzachanis, D.; et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: Phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet 2021, 398, 491–502. [Google Scholar] [CrossRef]
  177. Bishop, M.R.; Dickinson, M.; Purtill, D.; Barba, P.; Santoro, A.; Hamad, N.; Kato, K.; Sureda, A.; Greil, R.; Thieblemont, C.; et al. Second-Line Tisagenlecleucel or Standard Care in Aggressive B-Cell Lymphoma. N. Engl. J. Med. 2021, 386, 629–639. [Google Scholar] [CrossRef]
  178. Neelapu, S.S.; Dickinson, M.; Munoz, J.; Ulrickson, M.L.; Thieblemont, C.; Oluwole, O.O.; Herrera, A.F.; Ujjani, C.S.; Lin, Y.; Riedell, P.A.; et al. Axicabtagene ciloleucel as first-line therapy in high-risk large B-cell lymphoma: The phase 2 ZUMA-12 trial. Nat. Med. 2022, 28, 735–742. [Google Scholar] [CrossRef]
  179. Zhang, W.; Hu, B.; Jing, L.; Yang, J.; Wang, S.; Liu, Y.; Du, J.; Ren, Y.; Zhang, Y. Early Response Observed in Pediatric Patients with Refractory/Relapsed B-Cell Non-Hodgkin Lymphoma Treated with Sequential Chimeric Antigen Receptor T Cells. Blood 2019, 134 (Suppl. 1), 1945. [Google Scholar] [CrossRef]
  180. Minard, V.; Maude, S.L.; Buechner, J.; Krueger, J.; Locatelli, F.; Attarbaschi, A.; Laetsch, T.W.; Martínez, B.G.; Rubio, C.D.d.H.; Awasthi, R.; et al. Bianca: Phase II, single-arm, global trial to determine efficacy and safety of tisagenlecleucel in pediatric/young adult (YA) patients (Pts) with relapsed/refractory B-cell non-Hodgkin lymphoma (R/R B-NHL). J. Clin. Oncol. 2020, 38 (Suppl. 15), e22504. [Google Scholar] [CrossRef]
  181. Hill, L.; Lulla, P.; Heslop, H.E. CAR-T cell Therapy for Non-Hodgkin Lymphomas: A New Treatment Paradigm. Adv. Cell Gene Ther. 2019, 2, e54. [Google Scholar] [CrossRef] [PubMed]
  182. Kohorst, M.A.; Nunez, C.A.; Tewari, P.; Petropoulos, D.; Mahadeo, K.M.; Neelapu, S.S.; Shpall, E.J.; Khazal, S. Primary mediastinal large B-cell lymphoma in paediatric and adolescent patients: Emerging questions in the era of immunotherapy. Br. J. Haematol. 2020, 190, e114–e117. [Google Scholar] [CrossRef] [PubMed]
  183. Bouchkouj, N.; Zimmerman, M.; Kasamon, Y.L.; Wang, C.; Dai, T.; Xu, Z.; Wang, X.; Theoret, M.; Purohit-Sheth, T.; George, B. FDA Approval Summary: Axicabtagene Ciloleucel for Relapsed or Refractory Follicular Lymphoma. Oncologist 2022, 1–8. [Google Scholar] [CrossRef] [PubMed]
  184. Rosa, K. FDA Approves Liso-Cel for Refractory Large B-Cell Lymphoma. Available online: https://www.onclive.com/view/fda-approves-liso-cel-for-refractory-large-b-cell-lymphoma (accessed on 21 February 2022).
  185. Wang, L.; Li, L.-r.; Young, K.H. New agents and regimens for diffuse large B cell lymphoma. J. Hematol. Oncol. 2020, 13, 175. [Google Scholar] [CrossRef]
  186. Wayne, A.S.; Michel, G.; Lee, D.W.; Baruchel, A.; Chaudhury, S.; Brown, P.A.; Hermiston, M.; Krueger, J.; Shen, T.; Tailford, K.; et al. ZUMA-4: A Phase 1/2 Multicenter Study of KTE-X19 in Pediatric and Adolescent Patients with Relapsed/Refractory B Cell Acute Lymphoblastic Leukemia or Non-Hodgkin Lymphoma. Blood 2020, 136 (Suppl. 1), 42. [Google Scholar] [CrossRef]
  187. Wayne, A.S.; Huynh, V.; Hijiya, N.; Rouce, R.; Brown, P.A.; Krueger, J.; Kitko, C.L.; Ziga, E.D.; Hermiston, M.L.; Richards, M.K.; et al. ZUMA-4 Phase 1 Long-Term Results: KTE-X19 Chimeric Antigen Receptor T-Cell Therapy in Pediatric/Adolescent Patients with Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia. In Proceedings of the European Hematology Association Annual Meeting 2021, Virtual, 11 June 2021; European Hematology Association: The Hague, The Netherlands, 2021. Available online: https://onlinelibrary.wiley.com/doi/10.1002/pbc.27989 (accessed on 14 February 2022).
  188. Baird, J.H.; Frank, M.J.; Craig, J.; Patel, S.; Spiegel, J.Y.; Sahaf, B.; Oak, J.S.; Younes, S.F.; Ozawa, M.G.; Yang, E.; et al. CD22-directed CAR T-cell therapy induces complete remissions in CD19-directed CAR–refractory large B-cell lymphoma. Blood 2021, 137, 2321–2325. [Google Scholar] [CrossRef]
  189. Hu, Y.; Zhang, Y.; Zhao, H.; Wang, Y.; Nagler, A.; Chang, A.H.; Huang, H. CD19/CD22 Dual-Targeted Chimeric Antigen Receptor T-Cell Therapy for Relapsed/Refractory Aggressive B-Cell Lymphoma: A Safety and Efficacy Study. Blood 2020, 136 (Suppl. 1), 34. [Google Scholar] [CrossRef]
  190. Spiegel, J.Y.; Patel, S.; Muffly, L.; Hossain, N.M.; Oak, J.; Baird, J.H.; Frank, M.J.; Shiraz, P.; Sahaf, B.; Craig, J.; et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: A phase 1 trial. Nat. Med. 2021, 27, 1419–1431. [Google Scholar] [CrossRef]
  191. Deng, Q.; Han, G.; Puebla-Osorio, N.; Ma, M.C.J.; Strati, P.; Chasen, B.; Dai, E.; Dang, M.; Jain, N.; Yang, H.; et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat. Med. 2020, 26, 1878–1887. [Google Scholar] [CrossRef]
  192. Wang, Y.; Tong, C.; Dai, H.; Wu, Z.; Han, X.; Guo, Y.; Chen, D.; Wei, J.; Ti, D.; Liu, Z.; et al. Low-dose decitabine priming endows CAR T cells with enhanced and persistent antitumour potential via epigenetic reprogramming. Nat. Commun. 2021, 12, 409. [Google Scholar] [CrossRef]
  193. García-Domínguez, D.J.; Hontecillas-Prieto, L.; Palazón-Carrión, N.; Jiménez-Cortegana, C.; Sánchez-Margalet, V.; de la Cruz-Merino, L. Tumor Immune Microenvironment in Lymphoma: Focus on Epigenetics. Cancers 2022, 14, 1469. [Google Scholar] [CrossRef] [PubMed]
  194. Grywalska, E.; Rolinski, J. Epstein-Barr Virus–Associated Lymphomas. Semin. Oncol. 2015, 42, 291–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Bollard, C.M.; Gottschalk, S.; Torrano, V.; Diouf, O.; Ku, S.; Hazrat, Y.; Carrum, G.; Ramos, C.; Fayad, L.; Shpall, E.J.; et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J. Clin. Oncol. 2014, 32, 798–808. [Google Scholar] [CrossRef] [PubMed]
  196. Xu, J.; Niu, T. Natural killer cell-based immunotherapy for acute myeloid leukemia. J. Hematol. Oncol. 2020, 13, 167. [Google Scholar] [CrossRef] [PubMed]
  197. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef]
  198. Nato, Y.; Banerjee, P.; Kaur , I.; Thall, P.; Hosing, C.; Srour, S.A.; Griffin, L.; Barnett, M.; Champlin , R.E.; Shpall, E.J.; et al. Immunotherapy with ex vivo-expanded cord blood (CB)-derived nk cells combined with high-dose chemotherapy (HDC) and autologous stem cell transplant (ASCT) for B-cell non-hodgkin’s lymphoma (NHL). In Proceedings of the Transplant & Cellular Therapy Meetings; Available online: https://tct.confex.com/tct/2021/meetingapp.cgi/Paper/18012 (accessed on 8 February 2022).
  199. Robinson, S.; Boumendil, A.; Finel, H.; Khvedelidze, I.; Kanfer, E.; Peggs, K.; Furst, S.; Ram, R.; Marijt, W.; Vandenberghe, E.; et al. Donor Lymphocyte Infusions for the Treatment of Relapsed Non-Hodgkin Lymphoma Following Allogeneic Stem Cell Transplantation: A LWP-EBMT Study. Blood 2018, 132 (Suppl. 1), 707. [Google Scholar] [CrossRef]
  200. Kumar, A.; Watkins, R.; Vilgelm, A.E. Cell Therapy with TILs: Training and Taming T Cells to Fight Cancer. Front. Immunol. 2021, 12, 690499. [Google Scholar] [CrossRef]
  201. Rosenberg, S.A.; Yannelli, J.R.; Yang, J.C.; Topalian, S.L.; Schwartzentruber, D.J.; Weber, J.S.; Parkinson, D.R.; Seipp, C.A.; Einhorn, J.H.; White, D.E. Treatment of Patients with Metastatic Melanoma with Autologous Tumor-Infiltrating Lymphocytes and Interleukin 2. JNCI J. Nat. Cancer Inst. 1994, 86, 1159–1166. [Google Scholar] [CrossRef]
  202. Karyampudi, L.; Gokuldass, A.; Blaskovich, M.; Fardis, M.; Lotze, M.T. Phenotypic and functional characterization of tumor infiltrating lymphocytes (TIL) grown from non-hodgkin lymphoma tumors: Implications for the development of novel therapies for lymphoma. Ann. Oncol. 2017, 28, v363. [Google Scholar] [CrossRef]
  203. Tomassetti, S.; Chen, R.; Dandapani, S. The role of pembrolizumab in relapsed/refractory primary mediastinal large B-cell lymphoma. Ther. Adv. Hematol. 2019, 10, 2040620719841591. [Google Scholar] [CrossRef]
  204. Kwok, G.; Yau, T.C.; Chiu, J.W.; Tse, E.; Kwong, Y.L. Pembrolizumab (Keytruda). Hum. Vaccines Immunother. 2016, 12, 2777–2789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Armand, P.; Rodig, S.; Melnichenko, V.; Thieblemont, C.; Bouabdallah, K.; Tumyan, G.; Özcan, M.; Portino, S.; Fogliatto, L.; Caballero, M.D.; et al. Pembrolizumab in Relapsed or Refractory Primary Mediastinal Large B-Cell Lymphoma. J. Clin. Oncol. 2019, 37, 3291–3299. [Google Scholar] [CrossRef] [PubMed]
  206. Ansell, S.M.; Minnema, M.C.; Johnson, P.; Timmerman, J.M.; Armand, P.; Shipp, M.A.; Rodig, S.J.; Ligon, A.H.; Roemer, M.G.M.; Reddy, N.; et al. Nivolumab for Relapsed/Refractory Diffuse Large B-Cell Lymphoma in Patients Ineligible for or Having Failed Autologous Transplantation: A Single-Arm, Phase II Study. J. Clin. Oncol. 2019, 37, 481–489. [Google Scholar] [CrossRef]
  207. Geoerger, B.; Zwaan, C.M.; Marshall, L.V.; Michon, J.; Bourdeaut, F.; Casanova, M.; Corradini, N.; Rossato, G.; Farid-Kapadia, M.; Shemesh, C.S.; et al. Atezolizumab for children and young adults with previously treated solid tumours, non-Hodgkin lymphoma, and Hodgkin lymphoma (iMATRIX): A multicentre phase 1–2 study. Lancet Oncol. 2020, 21, 134–144. [Google Scholar] [CrossRef]
  208. Morris, S.W.; Kirstein, M.N.; Valentine, M.B.; Dittmer, K.G.; Shapiro, D.N.; Saltman, D.L.; Look, A.T. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin’s lymphoma. Science 1994, 263, 1281–1284. [Google Scholar] [CrossRef]
  209. Soda, M.; Choi, Y.L.; Enomoto, M.; Takada, S.; Yamashita, Y.; Ishikawa, S.; Fujiwara, S.; Watanabe, H.; Kurashina, K.; Hatanaka, H.; et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007, 448, 561–566. [Google Scholar] [CrossRef] [PubMed]
  210. Mossé, Y.P.; Voss, S.D.; Lim, M.S.; Rolland, D.; Minard, C.G.; Fox, E.; Adamson, P.; Wilner, K.; Blaney, S.M.; Weigel, B.J. Targeting ALK with Crizotinib in Pediatric Anaplastic Large Cell Lymphoma and Inflammatory Myofibroblastic Tumor: A Children’s Oncology Group Study. J. Clin. Oncol. 2017, 35, 3215–3221. [Google Scholar] [CrossRef] [PubMed]
  211. Pfizer’s Xalkori (Crizotinib) Approved by FDA for ALK-Positive Anaplastic Large Cell Lymphoma in Children and Young Adults [News Release]. Pfizer. Available online: https://www.pfizer.com/news/press-release/press-release-detail/pfizers-xalkorir-crizotinib-approved-fda-alk-positive (accessed on 16 February 2022).
  212. Hao, Y.; Chapuy, B.; Monti, S.; Sun, H.H.; Rodig, S.J.; Shipp, M.A. Selective JAK2 inhibition specifically decreases Hodgkin lymphoma and mediastinal large B-cell lymphoma growth in vitro and in vivo. Clin. Cancer Res. 2014, 20, 2674–2683. [Google Scholar] [CrossRef] [Green Version]
  213. Kim, S.J.; Yoon, D.H.; Kang, H.J.; Hong, J.Y.; Lee, H.S.; Oh, S.Y.; Shin, H.-J.; Kong, J.H.; Yi, J.H.; Sakamoto, K.; et al. Ruxolitinib shows activity against Hodgkin lymphoma but not primary mediastinal large B-cell lymphoma. BMC Cancer 2019, 19, 1080. [Google Scholar] [CrossRef] [Green Version]
Table 1. The molecular biology, genetics, and most common treatment regimens for common NHLs in the pediatric and adolescent population are described.
Table 1. The molecular biology, genetics, and most common treatment regimens for common NHLs in the pediatric and adolescent population are described.
NHL SubtypeMolecular Markers and Genetic AbnormalitiesTreatment Regimens
Burkitt lymphoma [12,62,63,64]CD19, CD20, immunoglobulin kappa or gamma light chains, IgM, CD10/CALLA
MYC translocations
TP53 mutation
t(8:14)(q24: q32), t(8;22)(q24;q11)
FAB/LMB Regimen:
COG-C5961 (FAB/LMB-96): intravenous (IV) formulations of cyclophosphamide (CPM), vincristine, prednisone/prednisolone, doxorubicin along with intrathecal (IT) methotrexate (MTX); IV Ifosfamide, etoposide, and MTX in higher risk group only (Group C)
Inter-B-NHL Ritux 2010: Addition of rituximab to the FAB/LMB backbone
BFM Regimen:
Involve IV formulation of Doxorubicin and CPM with doses lower than FAB/LMB; addition of ifosfamide, etoposide, and IV methotrexate including in lower and intermediate risk groups
Diffuse large B-cell lymphoma [62,63,64]Germinal center B-cell type: CD10 and BCL6
Activated B-cell type: MUM1
MYC locus translocations involving chromosome 8q24
Regimens similar to that used in BL
FAB:
COG-C5961 (FAB/LMB-96)
Inter-B-NHL Ritux 2010
BFM Regimen
Primary mediastinal large B-cell lymphoma [64,65]CD19, CD20, CD30, CD79
JAK2, STAT, PDL1/2 mutations
(DA-EPOCH-R): etoposide, doxorubicin, cyclophosphamide, vincristine, rituximab
Dismal response to FAB/LMB in comparison to responses seen in DLBCL (EFS of 66%)
No protocolized regimen
Anaplastic large cell lymphoma [62,66,67,68] CD30, t(2;5) translocations encoding NPM-ALKNHL-BFM studies: PO dexamethasone, IV methotrexate, IV etoposide, IV doxorubicin, IV vincristine, IV Vindesine and IT triple chemotherapy with methotrexate, cytarabine, and prednisone
APO Regimen: IV doxorubicin, PO prednisone, IV vincristine, PO 6-mercaptopurine and IT/IV methotrexate
Lymphoblastic lymphoma [45,69,70,71,72,73,74]B-cell type: CD19, cytoplasmic CD79a, cytoplasmic CD22 CD10, CD24, PAX5, CD20 and CD34 expression is variable
T Cell type: CD3, CD1a, CD2, CD4, CD5, CD7, CD8, CD10, CD44
COG AALL0434 (T-LL regimen): Augmented BFM regimen
IV Vincristine, IV Doxorubicin, IV Daunorubicin, IT MTX, IT Cytarabine, PO prednisone, IM peg-asparaginase, PO dexamethasone, IV/IM Cytarabine, Capizzi-style IV MTX (C-MTX), PO 6-mercaptopurine, PO thioguanine
C-MTX: escalating doses of IV MTX with initial dose of 100 mg/m2/day; no leucovorin rescue
NHL-BFM-95 (including B-type LL)
COG-A5971 (including B type LL): similar to COG AALL0434, except does not include C-MTX; use high dose MTX
Pediatric follicular lymphoma [52,62,75]Few cases with IRF4/MUM1 expression;
No widespread genetic abnormalities have been identified
NHL-BFM studies: 2–4 cycles of IV Doxorubicin, CPM, ifosfamide
Surgery in localized cases
R-CHOP
Abbreviations: Ara-C Cytarabine; BFM: Berlin–Frankfurt–Münster; CHOP: Cyclophosphamide–Doxorubicin–Vincristine–Prednisone; COG: Children’s Oncology Group; C-MTX: Capizzi-style IV Methotexate; CPM: Cyclophosphamide; DA-EPOCH-R: Etoposide–Prednisone–Vincristine–Cyclophosphamide–Doxorubicin–Rituximab; Dex: Dexamthasone; DNR: Daunorubicin; DXR: Doxorubicin; IE: Ifosfamide–Etoposide; IFO: Ifosfamide; IRF4: Interferon regulatory factor 4; IT: Intrathecal; IV: Intravenous; MTX: Methotrexate; PEG-ASP: PEG-Asparaginase; PO: oral; R-CHOP: Rituximab–Cyclophosphamide–Doxorubicin–Vincristine–Prednisone/prednisolone; TIT: Triple intrathecal therapy (MTX, Ara-C and Hydrocortisoine); VCR: Vincristine; VP-16: Etoposide; 6-MP: 6-mercaptopurine; 6-TG: Thioguanine.
Table 2. Recent advancements, including clinical trials and Food and Drug Administration (FDA) approved cellular and immunotherapies, in the treatment of pediatric and adolescent non-Hodgkin lymphoma are summarized below.
Table 2. Recent advancements, including clinical trials and Food and Drug Administration (FDA) approved cellular and immunotherapies, in the treatment of pediatric and adolescent non-Hodgkin lymphoma are summarized below.
Targeting Cell-Surface Molecules and DNA Replication
AgentClassTargetFDA Approval Key Clinical Trials in Pediatrics
RituximabmAbCD 20Approved for CD20+ DLBCL, BL, or mature B-ALL in pediatrics [105]NCT01516580; Inter-B-NHL Ritux 2010—phase 3 [106]
NCT00057811; ANHL01P1—phase 2 [107]
Brentuximab vedotin (BV)ADC conjugated to MMAECD30Approved for cHL and ALCL in adults
Not approved in pediatrics [108]
NCT01979536; ANHL12P1—phase 2 [109]
NCT02729961—phase 1/2 [110]
NCT02581631; CheckMate 436—phase 1/2 [111]
NCT03703050; NIVO-ALCL—phase 2 [112]
BlinatumomabBiTE CD19-CD3Approved for MRD + B-ALL after first or second remission and R/R B-ALL in adults and children [113]NCT00274742—phase 1 [114]
NCT01741792—phase 2 [115]
NCT03605589; Pembro-EB-1701—phase 1 [116]
NCT03117751; TOT17—phase 2/3 [117]
Inotuzumab ozogamicinADCCD22Approved for R/R B-ALL in adults
Not approved in pediatrics [118]
NCT02981628; AALL1621—phase 2 [119]; ITCC-059—phase 2 [120]
NelarabinePurine analogDNA cell synthesisApproved for R/R T-ALL and T-LL in adults and pediatrics [121]NCT00408005; AALL0434—phase 3 [122]
Cellular Therapy
TisagenlecleucelCAR T-cellCD19Approved for R/R B-ALL in pediatrics
R/R Large B Cell lymphoma in adults [123]
NCT03610724; BIANCA—phase 2 [124]
Axicabtagene CiloleucelCAR T-cellCD19Approved for
R/R Large B Cell lymphoma in adults
Not approved in pediatrics [125]
NCT02625480; ZUMA-4—phase 1/2 [126]
CD22 CAR T-cellsCAR T-cellCD22Not approvedNCT02315612—phase 1 [127]
NCT04088864—phase 1 [128]
CD19/CD22
CAR T-cells
CAR T-cellCD19/CD22Not approvedNCT03448393—phase 1 [129]
LMP1/2 CTLs (ALCI—Group A)Antigen-specific T cells(LMP) on EBV-associated NHLNot approvedNCT00062868; ALASCAR—phase 1 [130]
CAR-NK-CD19NK cellsCD19Not approvedNCT04796675; CAR-NK-CD19 cells—phase 1 [131]
Cord blood derived NK CellsNK cells Not approvedNCT03019640—phase 2 [132]
Donor lymphocyte infusion (DLI)DLIGVLNot approvedStudy by PPGHSCT [133]
Tumor-infiltrating Lymphocytes (TILs)TILsTumor specificNot approvedNone
Immune checkpoint inhibitors
PembrolizumabPD-1 inhibitorPD-1Approved for R/R cHL in adults and pediatrics
Approved for R/R PMBCL in adults and pediatrics [134]
NCT02332668; MK-3475-051/KEYNOTE-051—phase 1/2 [135]
NivolumabPD-1 inhibitorPD-1Approved for R/R cHL in adults
Not approved in pediatrics [136]
NCT02304458; ADVL1412—phase 1/2 [137]
NCT02927769; CheckMate 744—phase 2 [138]
AtezolizumabPDL-1 inhibitorPDL-1Not approved for adult or pediatric lymphoma [139]NCT02541604—phase 1/2 [140]
Targeted therapy
CrizotinibTKIALK-ROS1-MET pathwayALK-positive ALCL in pediatrics and young adults [141]NCT00939770; ADVL0912—phase 1/2 [142]
Abbreviations: ADC: Antibody Drug Conjugate; ALCL: Anaplastic Large Cell Lymphoma; ALK: Anaplastic Lymphoma Kinase; Axi-cel: Axicabtagene Ciloleucel; B-ALL; B-cell Acute Lymphoblastic Leukemia; BiTE: Bispecific T-Cell engager; BL: Burkitt Lymphoma; BLL: B Lymphoblastic Lymphoma; CAR T-cell: Chimeric Antigen Receptor T cell; CD: cluster of differentiation; cHL; Classical Hodgkin Lymphoma; CTL: Cytotoxic T Lymphocytes; DLBCL: Diffuse Large B-Cell Lymphoma; DLI: Donor Lymphocyte Infusion; EBV: Epstein Barr virus; GVL: Graft Versus Lymphoma; LMP: Latent Membrane Protein; mAb: monoclonal antibody; MMAE: Monomethyl auristatin E; MRD: Minimal residual Disease; NCT; National Clinical Trial; NK cells: Natural Killer Cells; PD-1: programmed cell death protein-1; PDL-1: programmed cell death protein ligand-1; PMBCL: Primary mediastinal large B-cell lymphoma; PPGHSCT: Polish Pediatric Group for Hematopoietic Stem Cell Transplantation; R/R: refractory/relapsed; T-ALL: T-cell Acute Lymphoblastic Leukemia; TILs: Tumor Infiltrating Lymphocytes; Tisa-cel: Tisagenlecleucel; TKI: Tyrosine Kinase Inhibitor; T-LL; T-cell Lymphoblastic Lymphoma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sheikh, I.N.; Elgehiny, A.; Ragoonanan, D.; Mahadeo, K.M.; Nieto, Y.; Khazal, S. Management of Aggressive Non-Hodgkin Lymphomas in the Pediatric, Adolescent, and Young Adult Population: An Adult vs. Pediatric Perspective. Cancers 2022, 14, 2912. https://doi.org/10.3390/cancers14122912

AMA Style

Sheikh IN, Elgehiny A, Ragoonanan D, Mahadeo KM, Nieto Y, Khazal S. Management of Aggressive Non-Hodgkin Lymphomas in the Pediatric, Adolescent, and Young Adult Population: An Adult vs. Pediatric Perspective. Cancers. 2022; 14(12):2912. https://doi.org/10.3390/cancers14122912

Chicago/Turabian Style

Sheikh, Irtiza N., Amr Elgehiny, Dristhi Ragoonanan, Kris M. Mahadeo, Yago Nieto, and Sajad Khazal. 2022. "Management of Aggressive Non-Hodgkin Lymphomas in the Pediatric, Adolescent, and Young Adult Population: An Adult vs. Pediatric Perspective" Cancers 14, no. 12: 2912. https://doi.org/10.3390/cancers14122912

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop