SlideShare uma empresa Scribd logo
1 de 13
Baixar para ler offline
1245
C H A P T E R 82
VIRUS-ASSOCIATED LYMPHOMA
Jennifer A. Kanakry and Richard F. Ambinder
There are five well-characterized human viruses that are generally
accepted as important in lymphomagenesis (Table 82-1). These
viruses may infect tumor cells (or their progenitors) or may act at a
distance. The genomes of Epstein-Barr virus (EBV), Kaposi sarcoma–
associated herpesvirus (KSHV, also known as human herpesvirus 8
[HHV-8]), and human T-lymphotropic virus-1 (HTLV-1) are present
in tumor cells. The viral genes expressed in tumor cells modulate
cellular metabolism, proliferation, and cell death. In contrast, the
human immunodeficiency virus (HIV) genome is generally not
detected in tumor cells. Whether hepatitis C virus (HCV) genomes
are present in lymphoma cells remains a subject of controversy.
Although viral infection plays a role in the pathogenesis of some
lymphomas, lymphomagenesis is unusual. Only a small subset of
infected people develops lymphoma. Furthermore, although primary
viral infection may be followed by lymphomagenesis within days or
weeks in exceptional circumstances, most lymphomas arise years or
decades after primary infection. Indeed the term adult in adult T-cell
leukemia/lymphoma (ATL) reflects the time lag between HTLV-1
infection in infancy and the evolution to malignancy. Geography and
associated environmental exposures, host genetic factors, and immune
status all modify risk.
Aspects of the biology and epidemiology of each of these viruses
and their relationship with lymphomagenesis are reviewed. In addi-
tion, clinically important and distinctive features of diagnosis and
treatment of the associated lymphomas are presented.
EPSTEIN-BARR VIRUS
Viral Biology
EBV is a gammaherpesvirus transmitted mainly through saliva.1,2
After primary infection, some of the infected cells are driven to pro-
liferate and thereby spread infection throughout the B-cell compart-
ment. Ultimately, in the normal host, there is an immune response
that controls infection and eradicates virus-infected proliferating
cells. Thereafter the viral genome is harbored mainly in resting
memory B lymphocytes that persist for life. These B cells that harbor
virus elude immune surveillance in part because of their very restricted
viral gene expression such that few viral antigens are presented. Occa-
sionally there is activation of viral lytic gene expression (at least in
some instances this occurs in concert with plasma cell differentiation)
leading to production of infectious virions that may infect other B
cells. T cell–mediated immune function keeps such proliferation in
check.3
In vitro EBV immortalizes B cells such that they grow indefinitely
as lymphoblastoid cell lines (LCLs) (Fig. 82-1). LCLs are tumorigenic
in immunodeficient mice. In LCL, viral genomes are present as cir-
cular double-stranded deoxyribonucleic acid (DNA) episomes within
the nucleus. The viral proteins required for immortalization include
Epstein-Barr virus nuclear antigen-1 (EBNA1), a sequence-specific
DNA-binding protein important in the maintenance of the viral
episome; EBNA2, a transcription factor that has many effects similar
to those of activated Notch receptors; and latent membrane protein-1
(LMP1), a constitutively activated member of the tumor necrosis
factor (TNF) receptor superfamily, which most closely resembles
CD40.4
LMP1 activates the nuclear factor kappa-B (NFκB) pathway,
which modulates cell proliferation and apoptosis.5
Several other EBV
proteins are also required for immortalization. Although EBV immor-
talization of B cells in vitro may offer some insights into tumorigen-
esis, some caution is required in using LCL as a tumor model. Most
EBV tumors, including tumors of B-lineage cells, do not express
many of the viral genes required for lymphocyte immortalization.
The only tumors that express the full complement of viral proteins
required for immortalization are those that arise in the most pro-
foundly immunocompromised patients (organ or hematopoietic
transplant recipients, patients with congenital immunodeficiency, or
patients with far advanced acquired immunodeficiency syndrome
[AIDS]). Thus in posttransplantation lymphoproliferative disorder
(PTLD), tumor cells may resemble LCL in expressing many viral
latency genes in association with normal karyotype (and few muta-
tions of the cellular genome). It has been suggested that there is an
inverse relationship between cellular mutations and viral gene expres-
sion in tumors.6
EBV gene expression may directly drive proliferation or inhibit
apoptotic pathways as illustrated by lymphocyte immortalization.
However, viral gene expression may also perturb normal lymphocyte
biology. Thus LMP1 expression upregulates activation induced (cyti-
dine) deaminase expression, which facilitates somatic hypermutation
and immunoglobulin class switching.7
LMP1 expression may also be
important in the conversion of naive B-cells to post-germinal center
memory B-cells. LMP2A allows B-cells that lack normal immuno-
globulin expression to escape regulatory checkpoints and survive.8
Epidemiology of Viral Infection
EBV infection is ubiquitous. The vast majority of adults are infected
worldwide. Primary infection is most often asymptomatic, especially
when it occurs in childhood.9
Primary infection may be associated
with the syndrome of infectious mononucleosis. Symptomatic
primary infection occurs more frequently in older children and in
adults than in younger children. Other possible determinants of
symptomatic primary infection include genetic factors and possibly
the size of the viral inoculum.
Strain differences in EBV are well recognized.10
However, the
importance of these strain differences with regard to lymphomagen-
esis remains poorly understood. There is general agreement that the
Type 1 strain EBV is most common worldwide and in tumors. The
Type 2 strain virus has been identified in some African Burkitt lym-
phoma (BL) and in some AIDS-associated lymphoma. A-strain virus
is more efficient at lymphocyte immortalization in vitro and lym-
phomagenesis in mouse models. The two strains of virus differ mainly
in the EBNA2 gene, but differences are recognized in some other
viral proteins as well.11
Variations in the regulatory regions or coding
regions of a variety of other genes including EBNA1, LMP1, and
ZTA have been recognized and suggested to play a role in
lymphomagenesis.
A simple classification of latent viral gene expression recognizes
three patterns as shown in the Table 82-2.
Part VII  Hematologic Malignancies1246
at very high copy number (perhaps millions of copies per cell) in
latently infected cells. The function(s) of these RNAs is disputed, but
their use for the detection of virus in a variety of surgical specimens
is generally accepted.
Viral antigens are detected by immunohistochemistry. In clinical
laboratories,immunohistochemistryforLMP1iscommonlyemployed
and is sensitive for the detection of EBV in Hodgkin lymphoma (HL).
In a variety of other EBV-associated B- andT-cell malignancies, expres-
sion is variable. Thus failure to detect LMP1 expression does not
exclude the presence of EBV except perhaps in HL. In principle, detec-
tion of EBNA1 should be universally applicable, although the low level
of antigen expression and the cross-reactivity of available monoclonal
antibodies have prevented immunohistochemistry for this antigen
from emerging as a standard tool.
Association With Particular Types of Lymphoma
Some lymphoma types are nearly 100% EBV associated, including
endemic BL, extranodal natural killer (NK)/T-cell lymphoma of the
nasal type, early PTLD, lymphomatoid granulomatosis, diffuse large
B-cell lymphoma (DLBCL) associated with chronic inflammation,
EBV-positive DLBCL of older adults, and AIDS primary central
nervous system (CNS) lymphoma (PCNSL).13-16
Other lymphoma
types are variably EBV associated. These include classic HL, PTLDs
occurring many months or years after transplantation, and systemic
AIDS-related lymphoma. Some lymphoma types appear never or
almost never to be EBV associated, including follicular lymphoma,
nodular lymphocyte-predominant HL, and mantle cell lymphoma.
Table 82-3 lists the lymphomas that have been associated with EBV,
associated cofactors, viral antigen expression, and an estimate of the
percentage of tumors within each lymphoma subtype that harbor
viral genomes.
Posttransplantation Lymphoproliferative Disorder
PTLD is a group of lymphoproliferative disorders ranging from poly-
clonal lymphoid hyperplasia to lymphomas that arise in patients after
solid organ or hematopoietic stem cell transplantation (HSCT).17
PTLD, especially in the first few months after transplantation, is
highly associated with EBV (Fig. 82-2, A, B). EBV gene expression
in PTLD corresponds to latencies 2 and 3.18
Broad expression of viral
proteins is seen only in immunosuppressed hosts, perhaps reflecting
that many of these proteins are commonly targeted by cytotoxic T
cells.
B cells that harbor EBV are able to proliferate in the setting of
posttransplantation immunosuppression at least in part due to
decreased T-cell surveillance.19
HSCT patients that receive grafts that
have been T-cell depleted develop EBV-associated PTLD at very high
rates. Treatment of rejection in solid organ transplant recipients with
agents such as the monoclonal antibody OKT3, which targets CD3+
cells, is associated with markedly increased risk for PTLD.20
Treat-
ment strategies such as the use of rituximab and infusion of EBV-
specific cytotoxic T cells have been quite effective in treating or
preventing PTLD (see box on Epstein-Barr Virus-Associated Positive
Posttransplant Lymphoproliferative Disorder).
Figure 82-1  EPSTEIN-BARR VIRUS (EBV)–IMMORTALIZED B CELL.
Normal B cells are readily immortalized in vitro with EBV. These cells express
EBV nuclear and membrane proteins. The nuclear proteins include a protein
expressed in all EBV-associated tumors, Epstein-Barr virus nuclear antigen 1
(EBNA1). This protein is required for episomal maintenance. Other viral
nuclear proteins expressed are transcription factors. These include EBNA-LP,
EBNA2, EBNA3A, EBNA3B, and EBNA3C. Two membrane proteins are
expressed: latent membrane protein 1 (LMP1), which activates NFκB path-
ways, and LMP2A, which mimics B-cell receptor (immunoglobulin)
signaling.
LMP2
LMP1
NF-κB
Cell
survival
B-cell receptor
EBNA1
Transcription
factors:
EBNA-LP
EBNA2
EBNA3A, EBNA3B,
EBNA3C
Table 82-1  Viruses and Lymphomagenesis
Virus Viral Genome in Tumor Cell Lymphoma Type
EBV Episomal B, T, NK
KSHV Episomal B
HTLV-1 Integrated T
HIV-1 Absent B
HCV Uncertain B
EBV, Epstein-Barr virus; HCV, hepatitis C virus; HIV-1, human
immunodeficiency virus type 1; HTLV-1, human T-lymphotropic virus-1; KSHV,
Kaposi sarcoma–associated herpesvirus; NK, natural killer.
Table 82-2  Patterns of Epstein-Barr Virus Gene Expression in Latency
Latency EBNA1
EBNA2, EBNA3A,
EBNA3B, EBNA3C LMP1 LMP2A
I +
II + + +
III + + + +
EBNA1, Epstein-Barr virus nuclear antigen 1; EBV, Epstein-Barr virus; LMP1,
latent membrane protein 1.
Epstein-Barr Virus Detection
in Clinical Specimens
The sensitivity of polymerase chain reaction (PCR) makes detection
of viral DNA straightforward, but the ubiquity of EBV infection, and
the persistence of B cells that harbor EBV in all seropositive individu-
als, means that EBV DNA is readily detected in many specimens that
include normal lymphocytes. Thus diagnosis of the EBV association
in general requires viral detection specifically in tumor cells. Tech-
niques for viral DNA detection by fluorescence in situ hybridization
or related techniques, although greatly improved in recent years,
remain the purview of research laboratories and are generally not
readily applicable to clinical specimens. This reflects the relatively low
copy number of the viral genome in tumor cells, typically 1 to 200
copies per cell. In contrast, in situ hybridization for the Epstein-Barr
encoded ribonucleic acids (RNAs) has emerged as a laboratory stan-
dard.12
These RNAs are polymerase 3 transcripts that are expressed
Chapter 82  Virus-Associated Lymphoma 1247
Hodgkin Lymphoma
Approximately 30% of HL tumors in the United States and Europe
are EBV associated (Fig. 82-2, C, D).21,22
Epidemiologic studies in
Denmark and Sweden suggest that individuals with a history of
symptomatic infectious mononucleosis are at increased risk for EBV-
associated HL, but not for EBV-negative HL or other lymphomas.23
The period of risk peaks at about 2 years but continues to be elevated
for at least 10 years after symptomatic mononucleosis.
Figure 82-2  EXAMPLES OF EPSTEIN-BARR VIRUS (EBV)–RELATED LYMPHOMAS. Posttransplantation lym-
phoproliferative disorder (PTLD), polymorphic type, EBV+ occurring in the gastrointestinal tract of a 15-month-old
girl following an orthotopic liver transplant (A and B). There was a mildly atypical lymphocytic infiltrate in the duodenal
mucosa composed of small lymphocytes, plasma cells, and occasional large cells (A). The infiltrate was EBV+ as dem-
onstrated by in situ hybridization for Epstein-Barr–encoded RNA (EBER) (B). Hodgkin lymphoma, nodular sclerosing
type, EBV+ (C and D). The hematoxylin-eosin section shows a portion of a lymph node from a cervical lymph node
biopsy of an 8-year-old girl. There are bands of sclerosis forming a cellular nodule, and within the nodule there is a
mixed inflammatory infiltrate and scattered large cells with contracted cytoplasm consistent with lacunar cells (C). The
immunophenotype of these cells was that of classic Hodgkin lymphoma, and the cells were EBER+ (D). EBV is seen
more frequently in mixed cellularity Hodgkin lymphoma but can be seen in 10% to 40% of cases of nodular sclerosing
type. It is even more frequent in cases associated with human immunodeficiency virus (HIV; see text) and in resource-
poor regions. Burkitt lymphoma, sporadic type, EBV+ (E and F). A section of the cervical lymph node biopsy of a
9-year-old girl with a rapidly enlarging neck mass is shown. The section illustrates the classic morphologic features of
Burkitt lymphoma with a “starry sky” appearance, sheets of intermediate-sized cells with multiple small nucleoli and
high mitotic rate. The cells were virtually all EBER+ (F). EBV can be seen in about 20% to 30% of cases of sporadic
Burkitt lymphoma and is essentially always positive in endemic cases.
F
A C E
B D F
A E
Table 82-3  Epstein-Barr Virus-Associated Lymphoma
Type Cofactors Viral Gene Expression Approximate % EBV Associated Comment
PTLD Immunosuppression,
allograft
Latency II or III 50-95 Early days/months after transplantation are more
commonly associated with EBV
Sporadic BL Latency I 20 in the United States Higher in Latin America
Endemic BL Malaria Latency I >95
AIDS BL HIV Latency I 30
HL Latency II 30 in the United States Higher % in mixed cellularity, in males, in Hispanics
AIDS PCNSL HIV Latency II or III >95
Nasal-type NK cell More common in Asia Latency II >95
AIDS PEL HIV and KSHV Latency I >75
AIDS, Acquired immunodeficiency syndrome; BL, Burkitt lymphoma; EBV, Epstein-Barr virus; HIV, human immunodeficiency virus; HL, Hodgkin lymphoma; KSHV,
Kaposi sarcoma–associated herpesvirus; NK, natural killer; PCNSL, primary central nervous system lymphoma; PEL, primary effusion lymphoma; PTLD,
posttransplantation lymphoproliferative disorder.
Higher EBV associations are seen in Latin America, Africa,
and parts of Asia. Factors associated with EBV tumor positivity
include mixed cellularity and lymphocyte-depleted classic HL histo-
logic subtypes, male gender, low socioeconomic background, history
of symptomatic infectious mononucleosis, and Hispanic ethnic-
ity.21,24,25
Organ and hematopoietic stem cell transplant recipients and
HIV-positive patients are more likely to develop HL than the general
population, and approximately 90% of the tumors are EBV
associated.26
Part VII  Hematologic Malignancies1248
Figure 82-3  EXAMPLES OF KSHV- AND HTLV-1-ASSOCIATED LYMPHOPROLIFERATIVE DISEASE.
Primary effusion lymphoma (A and B). A, The pleural tap had a high cell count, and the cytospin preparation revealed
a markedly pleomorphic cell population with large and giant cells with deep-blue cytoplasm. B, A cell block was pre-
pared (top) so that in situ hybridization studies could be performed. These studies showed the cells to be KSHV+ by
immunohistochemistry for latency associated nuclear antigen-1 (LANA-1) (bottom) and EBV+ by EBER in situ hybrid-
ization (not shown). C, Adult T-cell leukemia/lymphoma in a patient who was HTLV-1+. The peripheral smear showed
the classic “flower” cells. EBER, Epstein-Barr–encoded RNA; HTLV-1, human T-lymphotropic virus-1; KSHV, Kaposi
sarcoma–associated herpesvirus. (A and B courtesy Dr. Elizabeth Hyjek, University of Chicago.)
A B C
Epstein-Barr Virus-Associated Positive Posttransplant
Lymphoproliferative Disorder
A 55-year-old renal transplant patient presents with acute renal
failure 5 months after transplant. She is found on imaging to
have an obstructing mass in the transplanted kidney. She under-
goes kidney biopsy, and Epstein-Barr virus (EBV)–positive post-
transplant lymphoproliferative disorder (PTLD) involving the
transplanted organ is diagnosed. Treatment options include ritux-
imab; decreasing immunosuppression (acknowledging the associ-
ated risk for organ rejection); changing immunosuppressive
agents—switching a calcineurin inhibitor for a mammalian target
of rapamycin (mTOR) inhibitor; combination chemotherapy; or,
in the case of renal transplant, removal of the transplanted organ
and withdrawal of immunosuppression.
The EBV gene expression pattern in HL is latency II even when
HL occurs in immunocompromised populations.27
LMP1 and
LMP2A may mimic signaling of B-cell receptors and thus protect B
cells lacking functional immunoglobulin expression from apoptotic
signaling. Approximately 20% of HL lack productive immunoglobu-
lin gene rearrangements. These tumors appear to be exclusively EBV
associated.
The EBV association of HL appears not to have prognostic import
in young adult patients but is associated with poorer survival in older
patients in several reports.22
Treatment strategies involving adoptive
therapy with EBV-specific T cells expanded in vitro are under study.28
Burkitt Lymphoma
Endemic BL is nearly 100% associated with EBV, whereas sporadic
and HIV-associated BL are much more variably EBV associated (see
Table 82-3 and Fig. 82-2, E, F).29
Viral expression is latency I (i.e.,
EBNA1 is the only viral protein consistently expressed). The defining
feature of BL is a translocation between c-Myc on chromosome 8 and
one of the immunoglobulin genes on chromosomes 2, 14, or 22. It
has been generally presumed that falciparum malaria is a cofactor in
endemic BL, and the distribution of BL in Africa corresponds to the
distribution of holoendemic malaria. However, little is understood of
the pathogenesis or interaction between these infectious cofactors.
The characteristic presentation of BL is different in the endemic versus
sporadic settings, but there is no evidence to link these presentations
specifically with the virus. The virus-tumor association does not guide
diagnosis, therapy, or estimation of prognosis at present.
Diffuse Large B-cell Lymphoma Associated With
Chronic Inflammation
EBV-associated lymphomas sometimes arise in the setting of long-
standing chronic inflammation.15
This was first described in Japanese
patients with a remote history of pulmonary tuberculosis treated with
thoracoplasty with resulting chronic pyothorax, although many more
cases have since been reported. These patients developed EBV-
associated DLBCL of the pleural lining and associated lung tissue
decades after thoracoplasty. Similar cases of aggressive, EBV-associated
B-cell lymphomas have been reported to arise in patients at the sites
of chronic inflammation associated with various implants, surgical
mesh, or in the lung after chronic empyema.
KAPOSI SARCOMA–ASSOCIATED HERPESVIRUS
Virus and Tumor Epidemiology
KSHV (HHV-8) is a gammaherpesvirus that, unlike EBV, has a low
prevalence worldwide.30
The virus is endemic in certain areas, such
as in sub-Saharan Africa and the Middle East, and has an intermedi-
ate prevalence in Mediterranean countries. Transmission is believed
to be predominately through saliva. Similar to EBV, KSHV latently
infects B cells; viral genes that promote cell survival are implicated
in lymphomagenesis.
KSHV was discovered in Kaposi sarcoma but is also present in
two lymphoproliferative diseases: primary effusion lymphoma (PEL)
(Fig. 82-3, A, B) and multicentric Castleman disease (MCD).16,31
PEL
occurs almost exclusively in HIV-positive patients, particularly in
men who have sex with men, and typically when CD4 counts are less
than 100/mm3
. PELs are usually dually infected with KSHV and
EBV, but KSHV is always present. KSHV-associated MCD, although
much more common in HIV-infected populations, also occurs in the
general population.
MCD is a KSHV-associated, immunoglobulin M (IgM) λ–
producing nonclonal lymphoproliferative disorder typically involving
the mantle zone of lymph nodes and the spleen. The KSHV-positive
cells in MCD are always EBV negative. These cells express a broad
range of KSHV lytic antigens, and high KSHV copy numbers are
reported in plasma. Evolution into or coassociation with an aggressive
lymphoma, often of plasmablastic phenotype, is not uncommon.
Chapter 82  Virus-Associated Lymphoma 1249
HUMAN T-LYMPHOTROPIC VIRUS-1
Viral Biology
HTLV-1 is a retrovirus with a single-stranded RNA genome.37,38
Fol-
lowing infection there is reverse transcription and integration of
proviral DNA into the host genome. HTLV-1 infects a variety of cell
types but persists in CD4+
T lymphocytes. Viral infection within the
host is spread from cell to cell through direct cell-to-cell contact. As
with EBV, the proliferation of HTLV-1–infected lymphocytes plays
a central role in ensuring viral persistence.
The HTLV-1 protein Tax plays a central role in T-lymphocyte
immortalization and transformation (Fig. 82-4).37,39
Tax affects NFκB
and the serine/threonine kinase AKT pathways with diverse prolifera-
tive and antiapoptotic effects. As with some of the immunodominant
EBV antigens expressed in proliferating lymphocytes, Tax expression
is targeted by cytotoxic T cells. Another viral protein, Hbz, suppresses
Tax expression, allowing transformed T cells to elude immune sur-
veillance.40
Tax interferes with various DNA repair pathways and
induces reactive oxygen species, facilitating the development of aneu-
ploidy.37,39
Tax leads to functional inactivation of p53 and may inter-
fere with the spindle assembly checkpoint that normally operates in
mitosis to preserve euploidy.
Epidemiology of Viral Infection and Adult T-Cell
Leukemia/Lymphoma
HTLV-1 is endemic in particular regions of Japan, Africa, South
America, and some Caribbean islands.40,41
As assessed by seropreva-
lence, rates up to 37% are found on the southwestern Japanese islands
of Shikoku, Kyushu, and Okinawa, whereas most other areas of Japan
have an intermediate prevalence of 1% to 5%. In the United States
Within the HIV population, nearly all cases of MCD are KSHV
associated; this viral association is not as strong among HIV-negative
MCD patients. High expression of viral interleukin-6 (vIL-6) is
thought to contribute to the systemic inflammation seen in this
disorder.32
The KSHV-associated lymphomas that arise in association
with MCD are not nodal equivalents of PEL; rather these plasma-
blastic lymphomas are uniformly EBV negative and express IgM λ.16
Diagnostic and Therapeutic Considerations
The requisite finding in PEL is a lymphomatous effusion, which can
be pleural, pericardial, or peritoneal, without associated lymphade-
nopathy or masses, arising in the setting of immunocompromise.
Often, patients with HIV/AIDS will present with PEL in addition
to other KSHV-associated diseases, such as Kaposi sarcoma and
MCD, so thorough evaluation and staging should be undertaken at
diagnosis. On cytologic examination, the PEL tumor cells are large
with prominent nucleoli. The effusion cells are clonal B cells with
CD45 positivity but typically lack other specific B-cell lineage
markers, although CD30 and CD38 positivity can be seen. BCL6
mutations are frequently detected. Tumor cells always harbor KSHV
as demonstrated by staining for KSHV-associated latency-associated
nuclear antigen-1 (LANA-1). In MCD there is characteristically λ
light chain restriction.33
This does not reflect clonality as assessed by
study of Ig DNA rearrangements. Rather it reflects a tendency for
the virus to selectively infect cells expressing λ or to selectively drive
such cells to proliferate.
Treatment of PEL is most commonly combination chemotherapy.
Outcomes remain quite poor. Treatment of MCD with targeted
therapies has been more successful, including the use of rituximab
and IL-6 inhibition.34,35
Antivirals such as ganciclovir and valganci-
clovir have shown clinical activity in MCD, because viral lytic replica-
tion is a feature of this disease.36
Figure 82-4  HTLV-1 AND THE EVOLUTION OF ADULT T-CELL LEUKEMIA/LYMPHOMA. Following
HTLV-1 infection, many cells undergo apoptosis, but some infected CD4+
T cells are driven to proliferate by the
effects of Tax on the nuclear factor kappa-B (NFκB) pathway and on the AKT pathway. Tax is also suggested to
result in inactivation of p53, aneuploidy, and deoxyribonucleic acid (DNA) damage. Over many decades malignancy
evolves. HTLV-1, Human T-lymphotropic virus-1.
HTLV-1 infection
Tax activates AKT and NF-κB,
inactivates p53
Apoptosis
Tax
Tax
Tax
Tax
Viral control
by cytotoxic
T lymphocytes
CTL
Proliferation
Tax induces
aneuploidy
and DNA damage
30-40 years
Cellular
transformation
to ATL
Part VII  Hematologic Malignancies1250
infection may lead to cell death or establishment of latency in resting
cells. HIV infection is spread either through new rounds of virion
production with cellular infection or cell-cell fusion. There is no
evidence to suggest that infected cells are driven to proliferate. This
is in contrast to HTLV-1 and EBV, where proliferation of infected
cells appears to play a key role in establishing the long-term viral
reservoir and perhaps in mediating lymphomagenesis. The lympho-
mas that are increased in HIV-infected patients (Table 82-4) are of
B-cell lineage, and there is no substantial evidence that HIV infection
of B cells is important in the pathogenesis of these lymphomas.
Rather it appears that the HIV infection compromises cellular immu-
nity, decreasing immune surveillance of EBV- and KSHV-infected B
cells. In addition, HIV infection stimulates proliferation of B lym-
phocytes and perhaps genetic aberrations in the proliferating cells.51
Many possible mechanisms have been invoked, including (1) direct
stimulation of B cells by HIV antigens or antigens associated with
opportunistic infection; (2) stimulation of B cells by cellular proteins
(CD40 ligand) incorporated into the HIV virion leading to expres-
sion of activation-induced deaminase, an enzyme that mediates
double-strand DNA breaks; and (3) dysregulation of B cells as a
consequence of T-cell dysfunction.16,51,52
There is also some evidence that host biology may contribute to
lymphomagenesis. In particular, there are a variety of genetic poly-
morphisms that influence susceptibility to HIV-1 infection, such as
CCR5-Δ32.53,54
Individuals who are homozygous for this polymor-
phism are much less likely to be infected by HIV than others. Some
evidence has emerged to suggest that in heterozygotes who are
infected, HIV progression is slowed and there is less likelihood of
lymphomagenesis. Many of these lymphomas are associated with
EBV, KSHV, or both (see Table 82-4).
Epidemiology
Lymphoma is increased in all HIV risk groups, in contrast to Kaposi
sarcoma, which is rarely seen in injection drug users (or in hemo-
philiacs in an earlier era).55
There is a well-established relationship
between CD4+
cells per cubic millimeter overall and the risk for
lymphoma, but the relationship is complex and differs among lym-
phomas (see Table 82-4).
The incidence of non-Hodgkin lymphoma (NHL) in the HIV
population, particularly PCNSL (Fig. 82-5, A to C), has decreased
with the widespread use of highly active antiretroviral therapy
(HAART), although HIV patients on HAART still carry an increased
risk for lymphoma compared to the HIV-negative population.55
In
the HAART era, HIV patients on average have higher CD4+
counts
(often greater than 100 cells/mm3
) when diagnosed with lymphoma
as compared to the pre-HAART era. Among HIV patients with very
low CD4+
counts,56
NHLs are still seen at rates similar to the pre-
HAART era. Patients with HIV are now living much longer because
of effective antiretroviral regimens and decreased rates of opportunis-
the incidence in blood donors is 0.025%. It has been estimated that
about 20 million individuals are infected worldwide. The major
mode of transmission in endemic areas is from mother to child in
breast milk, although infection is also transmitted through sexual
intercourse, transfusion of cellular blood products, and the sharing
of needles and syringes. Evidence has been presented that HTLV-1
infection persists in association with certain human leukocyte antigen
(HLA) types and may be more readily transmitted from mother to
child when these HLA types are shared.
ATL is more common in men than in women and typically pres-
ents in the fourth or fifth decade of life. Perhaps as a function of the
long latency period, cases of ATL following blood transfusion or
needle sharing are vanishingly rare. The lifetime risk for ATL has been
estimated to be in excess of 6% in men who are HTLV-1 carriers in
an endemic region of Japan, although in other settings the risk may
be much lower.42
As with EBV, the subset of the infected population
that develops lymphoma is quite small.
Adult T-Cell Leukemia/Lymphoma
Diagnostic Considerations
There is a spectrum of HTLV-1–associated leukemia/lymphoma.38
Twenty percent of ATL cases present with a lymphomatous subtype
dominated by lymphadenopathy and hepatosplenomegaly. Cutane-
ous infiltration, lytic bone lesions, malignant effusions, and involve-
ment of the CNS and other extranodal sites are not uncommon.40
Hypercalcemia is common, particularly in leukemic forms. Presenta-
tions reflecting immune dysfunction such as strongyloidiasis with
dissemination, Pneumocystis jiroveci pneumonia (PJP), mycobacte-
rium, or cryptococcal infection are common. In the leukemic subtype
of ATL, the classic findings on peripheral blood smear are lympho-
cytes with flower-shaped nuclei (Fig. 82-3, C).
Histologically ATL shows lymph node effacement by large, atypi-
cal T cells usually expressing CD4+
, CD25+
, and CD52+
, with vari-
able CD30 and CD15 expression. Aneuploidy is consistent, although
characteristic cytogenetic abnormalities have not been identified.
Serologic analysis confirms the presence of HTLV-1 infection.38
Therapies Specific to HTLV-1 ATL
ATL is an aggressive lymphoma with poor responses to chemother-
apy, high relapse rates, and low overall survival. More indolent
chronic and smoldering forms are also recognized. Combination
chemotherapy regimens used in the treatment of other lymphomas
or leukemias are commonly used, but no standard regimen has
emerged.38,43
Several antivirals used in the treatment of HIV infection
have activity against HTLV-1. Among them are zidovudine and lami-
vudine. The combination of interferon and zidovudine has yielded
promising results, particularly in the leukemic subtype.38
Proteasome
and histone deacetylase (HDAC) inhibitors have attracted interest.
Given high CD25 and CD52 expression in ATL, the efficacy of
monoclonal antibodies aimed against these two receptors has been
investigated. Thus far alemtuzumab, an anti-CD52 monoclonal anti-
body, may have some activity in ATL based on small phase II studies
and case reports.44,45
Arsenic trioxide combined with interferon-α has
been shown to induce remissions in relapsed, refractory patients with
ATL, although durable responses were limited. In a trial of 20 ATL
patients, all-trans retinoic acid was used with 40% achieving remis-
sion. Allogeneic hematopoietic transplantation has been increasingly
recognized as an effective therapy for ATL.46,47
HIV-ASSOCIATED LYMPHOMAS
Viral Biology and Pathogenesis
HIV-1 is a retrovirus that infects CD4+
T cells and monocytes.48,49
It
appears to establish a lifelong reservoir in CD4+
T cells.50
Viral
Table 82-4  HIV-Associated Lymphoma
Lymphoma
CD4 Association
(cells/mm3
)
EBV Association
(%) Other Cofactors
PCNSL <50 >95
DLBCL Variable 40
BL >100 30
HL >100 90
PEL <100 >75 KSHV
BL, Burkitt lymphoma; DLBCL, diffuse large B-cell lymphoma; EBV, Epstein-
Barr virus; HIV, human immunodeficiency virus; HL, Hodgkin lymphoma;
KSHV, Kaposi sarcoma–associated herpesvirus; PCNSL, primary central
nervous system lymphoma; PEL, primary effusion lymphoma.
Chapter 82  Virus-Associated Lymphoma 1251
Diagnostic Considerations Specific to Lymphoma
in Patients With HIV
Lymphomas in HIV-infected patients are more likely to present with
B symptoms such as fever and night sweats, and advanced stage,
including bone marrow, extranodal, and CNS involvement. Thus the
approach to diagnosis is somewhat different than the approach in the
HIV-negative patient.
Unexplained fever and sweats in an HIV-seropositive patient, even
in the absence of lymphadenopathy, are sufficient to warrant consid-
eration of HL. Patterns of disease involvement also differ in HIV-
infected patients. Contiguous spread so characteristic of classic HL
in other settings is less common in HIV HL. Bone marrow–only
presentations of HL are not uncommon (Fig. 82-5, D to G), and the
possibility of HL should be in the differential diagnosis even in the
absence of lymphadenopathy.
Patients with HIV-associated NHL have higher rates of extranodal
involvement, including bone marrow and CNS disease, as well as
higher stage disease and more aggressive tumors on average.72
There-
fore it is recommended that all HIV-seropositive patients with aggres-
sive NHL undergo a diagnostic lumbar puncture. The routine use of
CNS intrathecal chemotherapy prophylaxis in all HIV-seropositive
NHL patients is controversial but reasonable and typically done in
particularly high-risk patients, such as those with BL, marrow or
testicular involvement, or extranodal disease.
Imaging is often more difficult to interpret in HIV patients than
in other settings. Lymphadenopathy associated with HIV infection
or opportunistic infection is common, and the presumption that
enlarged lymph nodes reflect the presence of lymphoma in patients
with known lymphoma or history of lymphoma is not as safe as in
tic infection. As a result, malignancy has emerged as the major cause
of mortality in HIV populations with access to antiretroviral
therapy.57,58
With the widespread use of HAART, the incidence of HL in HIV-
seropositive patients has not declined; patients with CD4+
counts
between 150 and 199 cells/mm3
actually have higher risk for
HIV-associated HL than patients with CD4+
counts of less than
50 cells/mm3
.59
HIV Hodgkin Lymphoma
A 42-year-old human immunodeficiency virus (HIV)–positive
patient, on highly active antiretroviral therapy (HAART) with a
CD4 count of 324 cells/mm3
and undetectable viral load, pres-
ents with fever, weight loss, and a palpable axillary lymph node.
Hodgkin lymphoma (HL) is diagnosed on excisional biopsy, and
Epstein-Barr virus (EBV) positivity is demonstrated by Epstein-
Barr–encoded RNA (EBER) in the Reed-Sternberg tumor cells.
Although he lacks additional lymphadenopathy, a bone marrow
biopsy is performed, and it shows involvement by HL. He has
stage IVB disease. Pneumocystis jiroveci pneumonia prophylaxis
is started despite adequate CD4 count in anticipation of chemo-
therapy. His HAART regimen is reviewed for potential antiviral-
chemotherapy drug interactions. He receives six cycles of
full-dose, first-line chemotherapy and achieves a complete
remission.
Figure 82-5  EXAMPLES OF HUMAN IMMUNODEFICIENCY VIRUS (HIV)–RELATED LYMPHOMAS. Primary central nervous system (CNS)
lymphoma in HIV+ patients (A to C). Gross appearance (coronal section) of the brain from an autopsy of a 24-year-old HIV+ female patient with temporo-
parietal mass due to a primary CNS lymphoma (A). The patient died from uncal and cingulate herniation. Biopsy section from another patient showing a
perivascular infiltrate of large lymphoma cells. This is the typical pattern of involvement by CNS lymphoma (B). The cells were shown to be CD20+
B cells
and were EBER+ (C). A diagnosis can be made without biopsy when magnetic resonance imaging studies show characteristic features and EBV is demonstrated
in the cerebrospinal fluid by polymerase chain reaction (see box on AIDS Primary Central Nervous System Lymphoma). Hodgkin lymphoma extensively
involving the bone marrow in an HIV+ patient with stage IVB disease (D to G). The bone marrow biopsy was entirely replaced with Hodgkin lymphoma
associated with dense sclerosis (D). An EBER study shows scattered positive cells throughout the marrow (E), corresponding to the Hodgkin and Reed-Sternberg
cells (F), which were CD30+
as illustrated (G). Hodgkin lymphoma infrequently involves the bone marrow in HIV-negative cases but some HIV+ patients can
first present with extensive bone marrow disease (see box on HIV Hodgkin Lymphoma). (A courtesy Dr. Peter Pytel, University of Chicago.)
A
B CC E G
D F
Part VII  Hematologic Malignancies1252
comparable outcomes regardless of HIV status.71
In this study all
HIV-seropositive patients were required to be on HAART to enroll,
and the majority had CD4+
counts of greater than 200 cells/mm3
.
Granulocyte colony-stimulating factor support was used throughout
chemotherapy cycles, as well as PJP and other antimicrobial prophy-
laxis. Differences in induction-related mortality, duration of neutro-
penia, progression-free survival, or overall survival were not detected,
although HIV-seropositive patients did have significantly more severe
mucositis and infectious complications.
Dose
Most would agree that dose reduction is appropriate in patients with
low CD4+
count (<100 cells/mm3
), history of ongoing opportunistic
infection, performance status below 75%, or compromised organ
function. Some regimens begin with a 50% dose reduction in cyclo-
phosphamide dose for CD4+
count of less than 100 cells/mm3
with
a built-in dose escalation for the next cycle if well tolerated.64,68
Chemotherapy regimens of varying intensity depending on CD4+
count, performance status, and International Prognostic Index score
have been studied, including a comparison of low-dose CHOP to
standard-dose CHOP, with no differences found in overall survival
based on the intensity of the chemotherapy regimen.66
In patients
with very low CD4+
counts, low-dose chemotherapy is a reasonable
treatment option, while maintaining the possibility of long-term
disease-free survival in some.
Antiretroviral Therapy
A few key special issues in considering concurrent antiretroviral
and lymphoma therapy include concerns about shared toxicities,
drug-drug interactions, and risk for inability to comply with consis-
tent antiretroviral dosing. Zidovudine is myelosuppressive and can
exacerbate pancytopenia associated with lymphoma therapy. If
patients are already on a zidovudine-containing regimen, it is generally
possible to substitute an alternative regimen before the initiation of
lymphoma therapy. Many antiretroviral agents alter the metabolism
of drugs used in lymphoma treatment. This has been a particular
concern when infusional chemotherapy regimens are used, and some
investigators have chosen to stop chemotherapy before initiation of
such regimens.64
However, in trials involving infusional chemotherapy
that allowed patients already on a stable antiretroviral regimen to
remain on that regimen during lymphoma treatment, major problems
were not noted.68
Nausea and vomiting associated with chemotherapy
regimens may interfere with regular antiviral dosing. Intermittent
antiretroviral therapy raises concerns about the development of a
resistant strain of HIV. Because of the concern that initiation of anti-
retroviral therapy with cytotoxic chemotherapy might result in such
resistance, many recommend delaying initiation of antiretroviral
therapy until an appropriate regimen to control nausea and vomiting
is established. Stopping antiretroviral therapy also carries with it some
risks. When antiretroviral therapy includes drugs with different half-
lives, stopping treatment may result in the longest-lived agent being
present in the absence of other antiretroviral agents. This is particu-
larly an issue for long-lived nonnucleoside reverse transcriptase inhibi-
tors. Even a single dose of such agents in the absence of other
antiretroviral agents may lead to resistance to that class of agents.Thus
when an interruption of antiretroviral therapy is planned, specific
strategies have been advocated, including a “staggered stop” or a
change to a regimen with components that have similar half-lives.62
For patients already on antiretroviral treatment at the time of
lymphoma diagnosis, the particulars of the regimen should be con-
sidered during the pretreatment evaluation. Atazanavir and indinavir
are associated with hyperbilirubinemia as a result of UGT1A1 inhibi-
tion.62
This is an unconjugated hyperbilirubinemia similar to that
occurring with Gilbert syndrome. Elevated total bilirubin in such
patients is not indicative of hepatic involvement or other serious
hepatic dysfunction and should not guide decisions about
other settings. Positron emission tomography–computerized tomog-
raphy (PET-CT), although useful, must also be interpreted with
caution insofar as HIV infection itself, opportunistic infection, and
immune reconstitution following the initiation of antiretroviral
therapy are all associated with signal on metabolic imaging with
fluorodeoxyglucose.
CD4+
counts can help to guide evaluation insofar as CD4+
counts
of greater than 300 cells/mm3
are typically associated with BL or
HL,60
whereas these diagnoses would be unlikely in patients with very
low CD4+
counts of less than 50 cells/mm3
(see box on AIDS Primary
Central Nervous System Lymphoma).61
Treatment
Aggressive chemotherapy for HIV-associated lymphoma was initially
associated with morbidity and mortality related to immunocompro-
mise. A phase III randomized study identified a reduced-dose regimen
as preferable to standard dose.63
Lower doses were not associated with
higher lymphoma cure rates but were associated with less
chemotherapy-related morbidity and mortality. However, with the
evolution of supportive care, including PJP prophylaxis and neutro-
phil growth factors, tolerance of chemotherapy improved. With effec-
tive antiretroviral therapy, long-term outcomes improved as well.64,65
And with full-dose therapies, some evidence emerged to suggest that
stage-for-stage outcomes might be as good or better in patients with
HIV-associated lymphoma compared to those without HIV.66
At the
outset of therapy a series of questions must be addressed.
Regimen
Rituximab, cyclophosphamide, hydroxydaunomycin, vincristine, and
prednisone (R-CHOP); and rituximab, infusional etoposide, predni-
sone, vincristine, cyclophosphamide, and hydroxydaunomycin
(R-EPOCH) have emerged as standard regimens.67,68
As in the treat-
ment of DLBCL in patients without HIV infection, the value of
infusional chemotherapy remains controversial. A recent pooled ret-
rospective analysis favored the infusional regimen.69
However, insofar
as the trials were conducted sequentially (R-CHOP between 1998
and 2002, and R-EPOCH between 2002 and 2006), it is difficult to
exclude other factors such as the availability of better antiretroviral
agents, improvements in supportive care, or changes in the popula-
tion studied. However, there is general agreement that rituximab
improves outcome, at least in patients with CD4+
counts of greater
than 50 cells/mm3
.
BL typically requires more intensive treatment regimens than
those used for DLBCL.70
Thus there has been concern about using
these regimens in the HIV-seropositive population. A small retrospec-
tive study of cyclophosphamide, vincristine (Oncovin), doxorubicin,
and methotrexate (CODOX-M)/ifosfamide, etoposide, and cytara-
bine (IVAC) for BL that included 14 patients with HIV showed
HIV-seropositive patients to have similar progression-free survival,
overall survival, and complete response rates as compared to the HIV-
negative BL patients. In a prospective Spanish study, HIV-seropositive
and HIV-seronegative BL patients were treated with six cycles of
intensive chemotherapy and rituximab and were found to have
AIDS Primary Central Nervous System Lymphoma
In acquired immunodeficiency syndrome (AIDS) primary central
nervous system lymphoma (PCNSL), Epstein-Barr virus (EBV)
polymerase chain reaction of the cerebrospinal fluid is positive
approximately 90% of the time and rarely positive in patients
with AIDS but without PCNSL. When EBV is detected in the
cerebrospinal fluid of an AIDS patient, coupled with characteris-
tic magnetic resonance imaging findings, this is sufficient to
diagnose AIDS PCNSL without a confirmatory brain biopsy.
Chapter 82  Virus-Associated Lymphoma 1253
whether infection of B cells plays any role in this process. A lym-
phoma cell line that produces infectious HCV has been reported.
Even in the absence of infection of B cells, interaction of the HCV
E2 protein with CD81 on B cells may drive B-cell proliferation or
lower the threshold for other B-cell stimuli to drive proliferation. Ig
signaling may be activated by Ig-virus complexes, and Toll-like recep-
tor 7 signaling may be activated by viral RNA. Finally, it is noted
that E2 binding triggers expression of activation-induced deaminase,
an enzyme that is important in generating somatic hypermutation
and that has also been implicated in mediating mutations thought to
play a role in DLBCL lymphomagenesis.77
Epidemiology of Viral Infection
and Associated Lymphoma
The association between HCV and lymphoma was first recognized
in patients with HCV-associated type II mixed cryoglobulinemia, an
autoimmune extrahepatic manifestation of HCV infection.79
There
followed demonstration of an increased risk for certain subtypes of
B-cell lymphoma (marginal zone lymphoma [MZL], lymphoplasma-
cytic lymphoma and to a lesser extent DLBCL in HCV-infected
patients).80-83
For example, in Taiwan the rate of chronic HCV infec-
tion in patients with NHL was 11%, 10-fold higher than in the
general Taiwanese population.84
Among HCV-infected patients with
lymphoma, nodal and splenic MZL, but not mucosa-associated lym-
phoid tissue (MALT) lymphomas, were increased. The HCV-
lymphoma association is more apparent in some countries than
others, with the association being established most clearly in Italy and
Japan. A multitude of studies in regions or countries where HCV
infection is less prevalent have failed to identify any association with
lymphoma.85-87
Further evidence in support of an etiologic relationship comes
from studies in which successful treatment of HCV was followed by
lymphoma regression.88
The most dramatic illustration comes from
patients with splenic lymphoma with villous lymphocytes treated
with ribavirin and interferon.
Certain HCV genotypes may confer increased risk for NHL, with
genotypes 2a/III and 2b/IV seen more frequently in the HCV-
seropositive patients that develop NHL.
Diagnostic Considerations
In contrast to EBV-, KSHV-, or HTLV-1–associated tumors, there is
no established role for studies demonstrating HCV nucleic acid or
protein in tumor cells. Thus serologic study and measurement of
HCV copy number are the only tools available for inferring an asso-
ciation. We recommend checking HCV serologic characteristics in
all patients with B-cell lymphomas most commonly associated with
chronic HCV infection.89
In addition, screening patients with chronic
HCV for a monoclonal gammopathy and cryoglobulinemia may be
of benefit to identify patients at highest risk for malignant transfor-
mation. Elevated serum γ-globulin levels have been found to be a
predictor of NHL among patients with type II mixed cryoglobuline-
mia.90
In patients who are HCV seropositive, we evaluate HCV RNA
in plasma.
Therapy
In patients with indolent lymphomas and untreated HCV infection,
antiviral treatment may obviate the need for cytotoxic chemotherapy
and should be considered as an initial therapeutic strategy. The
studies showing that antiviral therapy may lead to regression of HCV-
associated lymphomas involved treatment with ribavirin and inter-
feron. In the last year new antiviral agents have become available,
notably protease inhibitors specific for the HCV protease. Likelihood
of response to older therapy is a function of viral genotype, host
genetics (IL28R polymorphisms play a critical role in response to
chemotherapy dose adjustments. Ritonavir inhibits the clearance of
midazolam, phenytoin, and voriconazole and other agents metabo-
lized by the cytochrome P-450 CYP3A4 pathway.
Supportive Care
Although PJP prophylaxis is recommended only when the CD4+
count is less than 200 cells/mm3
for HIV patients not receiving
cytotoxic chemotherapy, prophylaxis is universally recommended for
HIV patients receiving cytotoxic chemotherapy. The following are
also commonly used: fungal prophylaxis with fluconazole; herpes
simplex and varicella prophylaxis with acyclovir, valacyclovir, or fam-
ciclovir; quinolone prophylaxis when neutrophil counts fall below
1000 cells/mm3
; and granulocyte growth factors.
Bone Marrow Transplantation in HIV Patients
Autologous bone marrow transplant has been successful in HIV-
seropositive NHL patients, with these patients having adequate stem
cell mobilization, nonrelapse mortality rates comparable to those for
HIV-negative patients, count recovery within 2 weeks of stem cell
rescue, and maintained control of HIV viral loads and CD4+
counts
after high-dose chemotherapy.73,74
There have also been successful
reduced-intensity allogeneic bone marrow transplants in HIV-
seropositive patients, making the possibilities for treating HIV
patients with NHL even more vast, even in those patients with
chemotherapy-resistant disease (see box on Autologous Bone Marrow
Transplant in an HIV-Seropositive Hodgkin Lymphoma Patient).75
HEPATITIS C VIRUS
Viral Biology and B-Lymphocyte Proliferation
HCV is an enveloped positive-strand RNA virus.76,77
Infection
involves interactions between E2, a viral structural protein with two
hypervariable regions, and a cellular protein CD81 present on hepa-
tocytes and B lymphocytes. A polyprotein is translated from viral
RNA and is cleaved by cellular and viral proteases, including NS3,
to yield proteins required for viral replication. The RNA-dependent
RNA polymerase that replicates the viral genome lacks proofreading
capacity, thus generating genetic heterogeneity among viral progeny.
Viral replication occurs predominantly in the liver, but some evidence
suggests that B cells may also be infected.
Chronic infection can be associated with mixed cryoglobulinemia,
a systemic vasculitis that results from clonal expansion of B cells
producing an IgM autoantibody against IgG, leading to deposition
of immune complexes on endothelial surfaces, resulting in inflamma-
tion.78
Several hypotheses have been advanced with regard to how
HCV might drive B-cell proliferation. There is controversy as to
Autologous Bone Marrow Transplant in an HIV-Seropositive Hodgkin
Lymphoma Patient
A 38-year-old human immunodeficiency virus (HIV)–positive
patient with CD4+
count of 485 cells/mm3
is diagnosed with
classic Hodgkin lymphoma (HL) and treated with doxorubicin
(Adriamycin), bleomycin, vinblastine, and dacarbazine (ABVD),
achieving a complete remission. Two years later he presents with
retroperitoneal lymphadenopathy and is found on biopsy to have
relapsed HL. He is treated with salvage chemotherapy with com-
plete response, as well as good performance status and no active
infections. His HIV remains well controlled on antiretroviral
therapy. He is deemed an excellent candidate for high-dose
therapy and undergoes consolidation with autologous bone
marrow transplant.
Part VII  Hematologic Malignancies1254
Choi I, Tanosaki R, Uike N, et al: Long-term outcomes after hematopoietic
SCT for adult T-cell leukemia/lymphoma: Results of prospective trials.
Bone Marrow Transplant 46:116, 2011.
Chuang SS, Liao YL, Chang ST, et al: Hepatitis C virus infection is signifi-
cantly associated with malignant lymphoma in Taiwan, particularly with
nodal and splenic marginal zone lymphomas. J Clin Pathol 63:595, 2010.
Engels EA, Pfeiffer RM, Landgren O, et al: Immunologic and virologic
predictors of AIDS-related non-Hodgkin lymphoma in the highly active
antiretroviral therapy era. J Acquir Immune Defic Syndr 54:78, 2010.
Epeldegui M, Hung YP, McQuay A, et al: Infection of human B cells with
Epstein-Barr virus results in the expression of somatic hypermutation-
inducing molecules and in the accrual of oncogene mutations. Mol
Immunol 44:934, 2007.
Evens AM, Roy R, Sterrenberg D, et al: Post-transplantation lymphoprolifera-
tive disorders: Diagnosis, prognosis, and current approaches to therapy.
Curr Oncol Rep 12:383, 2010.
Giordano TP, Henderson L, Landgren O, et al: Risk of non-Hodgkin lym-
phoma and lymphoproliferative precursor diseases in US veterans with
hepatitis C virus. JAMA 297:2010, 2007.
Guech-Ongey M, Simard EP, Anderson WF, et al: AIDS-related Burkitt
lymphoma in the United States: What do age and CD4 lymphocyte pat-
terns tell us about etiology and/or biology? Blood 116:5600, 2010.
Heslop HE, Slobod KS, Pule MA, et al: Long-term outcome of EBV-specific
T-cell infusions to prevent or treat EBV-related lymphoproliferative disease
in transplant recipients. Blood 115:925, 2010.
Hishizawa M, Kanda J, Utsunomiya A, et al: Transplantation of allogeneic
hematopoietic stem cells for adult T-cell leukemia: A nationwide retrospec-
tive study. Blood 116:1369, 2010.
Ito M, Kusunoki H, Mochida K, et al: HCV infection and B-cell lymphoma-
genesis. Adv Hematol 2011:835314, 2011.
Jaffe ES, Campo E, Swerdlow SH, et al: The 2008 WHO classification of
lymphoid neoplasms and beyond: Evolving concepts and practical applica-
tions. Blood 117:5019, 2011.
Keegan TH, Glaser SL, Clarke CA, et al: Epstein-Barr virus as a marker of
survival after Hodgkin’s lymphoma: A population-based study. J Clin Oncol
23:7604, 2005.
Kelly GL, Rickinson AB: Burkitt lymphoma: Revisiting the pathogenesis of
a virus-associated malignancy. Hematology Am Soc Hematol Educ Program
2007:277, 2007.
Kitahata MM, Achenbach CJ, Saag MS: Age at cancer diagnosis
among persons with AIDS. Ann Int Med 154:642; author reply 643,
2011.
Levine AM: HIV-associated lymphoma. Blood 115:2986, 2010.
Libra M, Polesel J, Russo AE, et al: Extrahepatic disorders of HCV infection:
A distinct entity of B-cell neoplasia? Int J Oncol 36:1331, 2010.
Matsuoka M, Jeang KT: Human T-cell leukaemia virus type 1 (HTLV-1)
infectivity and cellular transformation. Nat Rev Cancer 7:270, 2007.
Matsuoka M, Jeang KT: Human T-cell leukemia virus type 1 (HTLV-1) and
leukemic transformation: Viral infectivity, Tax, HBZ and therapy. Onco-
gene 30:1379, 2011.
Moore PS, Chang Y: KSHV: Forgotten but not gone. Blood 117:6973, 2011.
Ratner L, Harrington W, Feng X, et al: Human T cell leukemia virus reactiva-
tion with progression of adult T-cell leukemia-lymphoma. PLoS One
4:e4420, 2009.
Rudek MA, Flexner C, Ambinder RF: Use of antineoplastic agents in patients
with cancer who have HIV/AIDS. Lancet Oncol 12:905, 2011.
Savoldo B, Goss JA, Hammer MM, et al: Treatment of solid organ transplant
recipients with autologous Epstein Barr virus-specific cytotoxic T lympho-
cytes (CTLs). Blood 108:2942, 2006.
Sparano JA, Lee JY, Kaplan LD, et al: Rituximab plus concurrent infusional
EPOCH chemotherapy is highly effective in HIV-associated B-cell non-
Hodgkin lymphoma. Blood 115:3008, 2010.
Uldrick TS, Polizzotto MN, Aleman K, et al: High-dose zidovudine plus
valganciclovir for Kaposi sarcoma herpesvirus-associated multicentric Cas-
tleman disease: A pilot study of virus-activated cytotoxic therapy. Blood
117:6977, 2011.
Vereide DT, Sugden B: Lymphomas differ in their dependence on Epstein-
Barr virus. Blood 117:1977, 2011.
For complete list of references log on to www.expertconsult.com.
protease inhibitors) and other factors. The field is rapidly evolving,
and colleagues with specific expertise in appropriate antiviral
approaches should be consulted.76
Rituximab has posed an interesting dilemma for the treatment of
patients with HCV and lymphoma. It has been reported that HCV
plasma RNA increases following rituximab treatment, and there is
certainly the possibility that elimination of B cells for 3 to 18 months
following treatment may compromise humoral responses to the evo-
lution of HCV quasispecies. However, in studies to date overall
survival is not inferior.91
Similarly, combination chemotherapy is safe
in patients with HCV infection.92
Rituximab is specifically recom-
mended for the treatment of HCV-associated cryoglobulinemia
(although as in the treatment of Waldenström macroglobulinemia, it
must be appreciated that the initial response to rituximab may be an
increase in the IgM paraprotein level, necessitating plasmapheresis).
Aspects of Therapy
With regards to the use of rituximab to treat B-cell lymphomas in
patients with HCV, overall survival is not inferior, although there do
appear to be increased rates of hepatotoxicity and rises in HCV viral
load during therapy. Similarly, combination chemotherapy is safe in
patients with HCV infection, although HCV RNA levels can rise
during treatment. Interestingly, patients with HCV and splenic MZL
have had regression of their tumors with treatment for HCV infection
with interferon-α and ribavirin, an effect not seen in HCV-negative
patients with splenic MZL treated with the same regimen.
FUTURE DIRECTIONS
In this chapter a variety of virus-associated lymphomas and lympho-
proliferative diseases have been reviewed. For most of these lympho-
mas, standard antiviral drugs do not have a role in treatment. There
are, however, several exceptions, and these are worth highlighting.
Antiviral therapy for HCV-associated splenic lymphoma with villous
lymphocytes is accepted as a standard approach and likely has a role
in the treatment of other HCV-associated indolent lymphomas. Simi-
larly, ganciclovir or valganciclovir appears to have a role in the man-
agement of MCD associated with KSHV in HIV patients. And of
course there is an established role for antiretroviral therapy in the
treatment of HIV patients with malignancy. There are virus-targeted
therapies that are broadly accepted as standard, including adoptive
immunotherapy with EBV-specific T cells for PTLD. The use of
targeted T cells also has promise in other settings, including EBV-
associated HL. Other virus-targeted therapies are being developed.
Some involve vaccination; others involve induction of viral genes in
tumor cells, rendering them more susceptible to pharmacologic treat-
ment. Finally, it may ultimately be possible to prevent some kinds of
lymphoma by preventing viral infection or altering the host response
to viral infection.
SUGGESTED READINGS
Balsalobre P, Diez-Martin JL, Re A, et al: Autologous stem-cell transplanta-
tion in patients with HIV-related lymphoma. J Clin Oncol 27:2192,
2009.
Barta SK, Lee JY, Kaplan LD, et al: Pooled analysis of AIDS malignancy
consortium trials evaluating rituximab plus CHOP or infusional EPOCH
chemotherapy in HIV-associated non-Hodgkin lymphoma. Cancer doi:
10.1002/cncr.26723. [Epub ahead of print], 2011.
Bazarbachi A, Suarez F, Fields P, et al: How I treat adult T-cell leukemia/
lymphoma. Blood 118:1736, 2011.
Bower M, Newsom-Davis T, Naresh K, et al: Clinical features and outcome
in HIV-associated multicentric Castleman’s disease. J Clin Oncol 29:2481,
2011.
Carbone A, Cesarman E, Spina M, et al: HIV-associated lymphomas and
gamma-herpesviruses. Blood 113:1213, 2009.
Chapter 82  Virus-Associated Lymphoma 1254.e1
Key Words
Adult T-cell leukemia/lymphoma
Burkitt lymphoma
Epstein-Barr virus
Hepatitis C virus
Hodgkin lymphoma
Human T-lymphotropic virus-1
Human immunodeficiency virus type 1
Kaposi sarcoma–associated herpesvirus (human herpesvirus 8)
Marginal zone lymphoma
Natural killer/T-cell lymphoma
Posttransplant lymphoma
Splenic lymphoma
Part VII  Hematologic Malignancies1254.e2
REFERENCES
1.	 Cohen JI: Epstein-Barr virus infection. N Engl J Med 343:481, 2000.
2.	 Thorley-Lawson DA, Gross A: Persistence of the Epstein-Barr virus and
the origins of associated lymphomas. N Engl J Med 350:1328, 2004.
3.	 Savoldo B, Goss JA, Hammer MM, et al: Treatment of solid organ
transplant recipients with autologous Epstein Barr virus-specific cyto-
toxic T lymphocytes (CTLs). Blood 108:2942, 2006.
4.	 Roughan JE, Torgbor C, Thorley-Lawson DA: Germinal center B cells
latently infected with Epstein-Barr virus proliferate extensively but do
not increase in number. J Virol 84:1158, 2010.
5.	 Mainou BA, Raab-Traub N: LMP1 strain variants: Biological and molec-
ular properties. J Virol 80:6458, 2006.
6.	 Vereide DT, Sugden B: Lymphomas differ in their dependence on
Epstein-Barr virus. Blood 117:1977, 2011.
7.	 Epeldegui M, Hung YP, McQuay A, et al: Infection of human B cells
withEpstein-Barrvirusresultsintheexpressionofsomatichypermutation-
inducing molecules and in the accrual of oncogene mutations. Mol
Immunol 44:934, 2007.
8.	 Rovedo M, Longnecker R: Epstein-Barr virus latent membrane protein
2A preferentially signals through the Src family kinase Lyn. J Virol
82:8520, 2008.
9.	 Williams H, Crawford DH: Epstein-Barr virus: The impact of scientific
advances on clinical practice. Blood 107:862, 2006.
10.	 Higgins CD, Swerdlow AJ, Macsween KF, et al: A study of risk factors
for acquisition of Epstein-Barr virus and its subtypes. J Infect Dis
195:474, 2007.
11.	 Tierney RJ, Edwards RH, Sitki-Green D, et al: Multiple Epstein-Barr
virus strains in patients with infectious mononucleosis: Comparison of
ex vivo samples with in vitro isolates by use of heteroduplex tracking
assays. J Infect Dis 193:287, 2006.
12.	 Glaser SL, Gulley ML, Borowitz MJ, et al: Inter- and intra-observer
reliability of Epstein-Barr virus detection in Hodgkin lymphoma using
histochemical procedures. Leuk Lymphoma 45:489, 2004.
13.	 Hsieh PP, Tung CL, Chan AB, et al: EBV viral load in tumor tissue is
an important prognostic indicator for nasal NK/T-cell lymphoma. Am J
Clin Patho 128:579, 2007.
14.	 MacMahon EM, Glass JD, Hayward SD, et al: Epstein-Barr virus in
AIDS-related primary central nervous system lymphoma. Lancet
338:969, 1991.
15.	 Jaffe ES, Campo E, Swerdlow SH, et al: The 2008 WHO classification
of lymphoid neoplasms and beyond: Evolving concepts and practical
applications. Blood 117:5019, 2011.
16.	 Carbone A, Cesarman E, Spina M, et al: HIV-associated lymphomas and
gamma-herpesviruses. Blood 113:1213, 2009.
17.	 Evens AM, Roy R, Sterrenberg D, et al: Post-transplantation lympho­
proliferative disorders: Diagnosis, prognosis, and current approaches to
therapy. Curr Oncol Rep 12:383, 2010.
18.	 Murray PG, Deacon E, Young LS, et al: Localization of Epstein-Barr
virus in Castleman’s disease by in situ hybridization and immunohisto-
chemistry. Hematol Pathol 9:17, 1995.
19.	 Heslop HE, Slobod KS, Pule MA, et al: Long-term outcome of EBV-
specific T-cell infusions to prevent or treat EBV-related lymphoprolifera-
tive disease in transplant recipients. Blood 115:925, 2010.
20.	 Swinnen LJ, Costanzo-Nordin MR, Fisher SG, et al: Increased incidence
of lymphoproliferative disorder after immunosuppression with the
monoclonal antibody OKT3 in cardiac-transplant recipients. N Engl J
Med 323:1723, 1990.
21.	 Glaser SL, Lin RJ, Stewart SL, et al: Epstein-Barr virus-associated Hodg-
kin’s disease: Epidemiologic characteristics in international data. Int J
Cancer 70:375, 1997.
22.	 Keegan TH, Glaser SL, Clarke CA, et al: Epstein-Barr virus as a marker
of survival after Hodgkin’s lymphoma: A population-based study. J Clin
Oncol 23:7604, 2005.
23.	 Hjalgrim H, Askling J, Rostgaard K, et al: Characteristics of Hodgkin’s
lymphoma after infectious mononucleosis. N Engl J Med 349:1324,
2003.
24.	 Ambinder RF, Browning PJ, Lorenzana I, et al: Epstein-Barr virus and
childhood Hodgkin’s disease in Honduras and the United States. Blood
81:462, 1993.
25.	 Glaser SL, Gulley ML, Clarke CA, et al: Racial/ethnic variation in EBV-
positive classical Hodgkin lymphoma in California populations. Int J
Cancer 123:1499, 2008.
26.	 Glaser SL, Clarke CA, Gulley ML, et al: Population-based patterns of
human immunodeficiency virus-related Hodgkin lymphoma in the
Greater San Francisco Bay Area, 1988-1998. Cancer 98:300, 2003.
27.	 Siebert JD, Ambinder RF, Napoli VM, et al: Human immunodeficiency
virus-associated Hodgkin’s disease contains latent, not replicative,
Epstein-Barr virus. Hum Pathol 26:1191, 1995.
28.	 Bollard CM, Aguilar L, Straathof KC, et al: Cytotoxic T lymphocyte
therapy for Epstein-Barr virus+ Hodgkin’s disease. J Exp Med 200:1623,
2004.
29.	 Kelly GL, Rickinson AB: Burkitt lymphoma: Revisiting the pathogenesis
of a virus-associated malignancy. Hematology Am Soc Hematol Educ
Program 2007:277, 2007.
30.	 Moore PS, Chang Y: KSHV: Forgotten but not gone. Blood 117:6973,
2011.
31.	 Ambinder RF, Cesarman E: Clinical and pathological aspects of EBV
and KSHV infection. In: Arvin A, Campadelli-Fiume G, Mocarski E,
et al, editors: Human herpesviruses: Biology, therapy, and immunoprophy-
laxis, Cambridge, 2007, Cambridge University Press.
32.	 Cannon JS, Nicholas J, Orenstein JM, et al: Heterogeneity of viral
IL-6 expression in HHV-8-associated diseases. J Infect Dis 180:824,
1999.
33.	 Du MQ, Liu H, Diss TC, et al: Kaposi sarcoma-associated herpesvirus
infects monotypic (IgM lambda) but polyclonal naive B cells in Castle-
man disease and associated lymphoproliferative disorders. Blood 97:2130,
2001.
34.	 Bower M, Newsom-Davis T, Naresh K, et al: Clinical features and
outcome in HIV-associated multicentric Castleman’s disease. J Clin
Oncol 29:2481, 2011.
35.	 Casper C: The aetiology and management of Castleman disease at 50
years: Translating pathophysiology to patient care. Br J Haematol 129:3,
2005.
36.	 Uldrick TS, Polizzotto MN, Aleman K, et al: High-dose zidovudine plus
valganciclovir for Kaposi sarcoma herpesvirus-associated multicentric
Castleman disease: A pilot study of virus-activated cytotoxic therapy.
Blood 117:6977, 2011.
37.	 Matsuoka M, Jeang KT: Human T-cell leukemia virus type 1 (HTLV-1)
and leukemic transformation: Viral infectivity, Tax, HBZ and therapy.
Oncogene 30:1379, 2011.
38.	 Bazarbachi A, Suarez F, Fields P, et al: How I treat adult T-cell leukemia/
lymphoma. Blood 118:1736, 2011.
39.	 Bellon M, Baydoun HH, Yao Y, et al: HTLV-I Tax-dependent and
-independent events associated with immortalization of human primary
T lymphocytes. Blood 115:2441, 2010.
40.	 Matsuoka M, Jeang KT: Human T-cell leukaemia virus type 1 (HTLV-1)
infectivity and cellular transformation. Nat Rev Cancer 7:270, 2007.
41.	 Sonoda S, Li HC, Tajima K: Ethnoepidemiology of HTLV-1 related
diseases: Ethnic determinants of HTLV-1 susceptibility and its world-
wide dispersal. Cancer Sci 102:295, 2011.
42.	 Koga Y, Iwanaga M, Soda M, et al: Trends in HTLV-1 prevalence and
incidence of adult T-cell leukemia/lymphoma in Nagasaki, Japan. J Med
Virol 82:668, 2010.
43.	 Ratner L, Harrington W, Feng X, et al: Human T cell leukemia virus
reactivation with progression of adult T-cell leukemia-lymphoma. PLoS
One 4:e4420, 2009.
44.	 Jiang L, Yuan CM, Hubacheck J, et al: Variable CD52 expression in
mature T cell and NK cell malignancies: Implications for alemtuzumab
therapy. Br J Haematol 145:173, 2009.
45.	 Morris J, Sharma K, O’Mahony D, et al: A phase II study of the efficacy
and toxicity of alemtuzumab (Campath) for treatment of human T cell
lymphotrophic virus-1 (HTLV-1)-associated adult T cell leukemia/
lymphoma (ATL). AIDS Res Hum Retroviruses 23:632, 2007.
46.	 Choi I, Tanosaki R, Uike N, et al: Long-term outcomes after hemato-
poietic SCT for adult T-cell leukemia/lymphoma: Results of prospective
trials. Bone Marrow Transplant 46:116, 2011.
47.	 Hishizawa M, Kanda J, Utsunomiya A, et al: Transplantation of alloge-
neic hematopoietic stem cells for adult T-cell leukemia: A nationwide
retrospective study. Blood 116:1369, 2010.
Chapter 82  Virus-Associated Lymphoma 1254.e3
48.	 Epeldegui M, Vendrame E, Martinez-Maza O: HIV-associated immune
dysfunction and viral infection: Role in the pathogenesis of AIDS-related
lymphoma. Immunol Res 48:72, 2010.
49.	 Kitahata MM, Achenbach CJ, Saag MS: Age at cancer diagnosis among
persons with AIDS. Ann Intern Med 154:642; author reply 643, 2011.
50.	 Shen L, Siliciano RF: Viral reservoirs, residual viremia, and the potential
of highly active antiretroviral therapy to eradicate HIV infection. J Allergy
Clin Immunol 122:22, 2008.
51.	 Gaidano G, Pasqualucci L, Capello D, et al: Aberrant somatic hypermu-
tation in multiple subtypes of AIDS-associated non-Hodgkin lymphoma.
Blood 102:1833, 2003.
52.	 Epeldegui M, Thapa DR, De la Cruz J, et al: CD40 ligand (CD154)
incorporated into HIV virions induces activation-induced cytidine
deaminase (AID) expression in human B lymphocytes. PloS one 5:e11448,
2010.
53.	 Dean M, Jacobson LP, McFarlane G, et al: Reduced risk of AIDS lym-
phoma in individuals heterozygous for the CCR5-Δ32 mutation. Cancer
Res 59:3561, 1999.
54.	 Goedert JJ: The epidemiology of acquired immunodeficiency syndrome
malignancies. Semin Oncol 27:390, 2000.
55.	 Levine AM: HIV-associated lymphoma. Blood 115:2986, 2010.
56.	 Bohlius J, Schmidlin K, Costagliola D, et al: Incidence and risk factors
of HIV-related non-Hodgkin’s lymphoma in the era of combination
antiretroviral therapy: A European multicohort study. Antivir Ther
14:1065, 2009.
57.	 Bonnet F, Lewden C, May T, et al: Malignancy-related causes of death
in human immunodeficiency virus-infected patients in the era of highly
active antiretroviral therapy. Cancer 101:317, 2004.
58.	 Engels EA, Pfeiffer RM, Landgren O, et al: Immunologic and virologic
predictors of AIDS-related non-Hodgkin lymphoma in the highly active
antiretroviral therapy era. J Acquir Immune Defic Syndr 54:78, 2010.
59.	 Biggar RJ, Jaffe ES, Goedert JJ, et al: Hodgkin lymphoma and immu-
nodeficiency in persons with HIV/AIDS. Blood 108:3786, 2006.
60.	 Guech-Ongey M, Simard EP, Anderson WF, et al: AIDS-related Burkitt
lymphoma in the United States: What do age and CD4 lymphocyte
patterns tell us about etiology and/or biology? Blood 116:5600, 2010.
61.	 Ambinder RF, Bhatia K, Martinez-Maza O, et al: Cancer biomarkers in
HIV patients. Curr Opin HIV AIDS 5:531, 2010.
62.	 Rudek MA, Flexner C, Ambinder RF: Use of antineoplastic agents in
patients with cancer who have HIV/AIDS. Lancet Oncol 12:905, 2011.
63.	 Kaplan LD, Straus DJ, Testa MA, et al: Low-dose compared with
standard-dose m-BACOD chemotherapy for non-Hodgkin’s lymphoma
associated with human immunodeficiency virus infection. National Insti-
tute of Allergy and Infectious Diseases AIDS Clinical Trials Group.
N Engl J Med 336:1641, 1997.
64.	 Little RF, Pittaluga S, Grant N, et al: Highly effective treatment of
acquired immunodeficiency syndrome-related lymphoma with dose-
adjusted EPOCH: Impact of antiretroviral therapy suspension and
tumor biology. Blood 101:4653, 2003.
65.	 Ratner L, Lee J, Tang S, et al: Chemotherapy for human immunodefi-
ciency virus-associated non-Hodgkin’s lymphoma in combination with
highly active antiretroviral therapy. J Clin Oncol 19:2171, 2001.
66.	 Mounier N, Spina M, Gabarre J, et al: AIDS-related non-Hodgkin
lymphoma: Final analysis of 485 patients treated with risk-adapted inten-
sive chemotherapy. Blood 107:3832, 2006.
67.	 Kaplan LD, Lee JY, Ambinder RF, et al: Rituximab does not improve
clinical outcome in a randomized phase 3 trial of CHOP with or without
rituximab in patients with HIV-associated non-Hodgkin lymphoma:
AIDS-Malignancies Consortium Trial 010. Blood 106:1538, 2005.
68.	 Sparano JA, Lee JY, Kaplan LD, et al: Rituximab plus concurrent infu-
sional EPOCH chemotherapy is highly effective in HIV-associated B-cell
non-Hodgkin lymphoma. Blood 115:3008, 2010.
69.	 Barta SK, Lee JY, Kaplan LD, et al: Pooled analysis of AIDS malignancy
consortium trials evaluating rituximab plus CHOP or infusional
EPOCH chemotherapy in HIV-associated non-Hodgkin lymphoma.
Cancer doi: 10.1002/cncr.26723. [Epub ahead of print], 2011.
70.	 Levine AM: Management of AIDS-related lymphoma. Curr Opin Oncol
20:522, 2008.
71.	 Oriol A, Ribera JM, Bergua J, et al: High-dose chemotherapy and immu-
notherapy in adult Burkitt lymphoma: Comparison of results in human
immunodeficiency virus-infected and noninfected patients. Cancer
113:117, 2008.
72.	 Gopal S, Martin K, Richards K, et al: Clinical presentation, treatment,
and outcomes among 65 patients with HIV-associated lymphoma treated
at the University of North Carolina, 2000-2010. AIDS Res Hum Retro-
viruses [Epub ahead of print] 2011.
73.	 Balsalobre P, Diez-Martin JL, Re A, et al: Autologous stem-cell trans-
plantation in patients with HIV-related lymphoma. J Clin Oncol
27:2192, 2009.
74.	 Gabarre J, Marcelin AG, Azar N, et al: High-dose therapy plus autolo-
gous hematopoietic stem cell transplantation for human immunodefi-
ciency virus (HIV)-related lymphoma: Results and impact on HIV
disease. Haematologica 89:1100, 2004.
75.	 Bryant A, Milliken S: Successful reduced-intensity conditioning alloge-
neic HSCT for HIV-related primary effusion lymphoma. Biol Blood
Marrow Transplant 14:601, 2008.
76.	 Rosen HR: Clinical practice. Chronic hepatitis C infection. N Engl
J Med 364:2429, 2011.
77.	 Ito M, Kusunoki H, Mochida K, et al: HCV infection and B-cell lym-
phomagenesis. Adv Hematol 2011:835314, 2011.
78.	 Della Rossa A, Baldini C, Tavoni A, et al: How HCV has changed the
approach to mixed cryoglobulinemia. Clin Exp Rheumatol 27:S115,
2009.
79.	 Monti G, Pioltelli P, Saccardo F, et al: Incidence and characteristics of
non-Hodgkin lymphomas in a multicenter case file of patients with
hepatitis C virus-related symptomatic mixed cryoglobulinemias. Arch
Intern Med 165:101, 2005.
80.	 Anderson LA, Engels EA: Hepatitis C virus infection and non-Hodgkin
lymphoma: Interesting association or causal relationship? Int J Cancer
122:x, 2008.
81.	 Giordano TP, Henderson L, Landgren O, et al: Risk of non-Hodgkin
lymphoma and lymphoproliferative precursor diseases in US veterans
with hepatitis C virus. JAMA 297:2010, 2007.
82.	 Libra M, Polesel J, Russo AE, et al: Extrahepatic disorders of HCV infec-
tion: A distinct entity of B-cell neoplasia? Int J Oncol 36:1331, 2010.
83.	 Amin J, Dore GJ, O’Connell DL, et al: Cancer incidence in people with
hepatitis B or C infection: A large community-based linkage study. J
Hepatol 45:197, 2006.
84.	 Chuang SS, Liao YL, Chang ST, et al: Hepatitis C virus infection is
significantly associated with malignant lymphoma in Taiwan, particularly
with nodal and splenic marginal zone lymphomas. J Clin Pathol 63:595,
2010.
85.	 Varma S, Menon MC, Garg A, et al: Hepatitis C virus infection among
patients with non-Hodgkin’s lymphoma in northern India. Hepatol Int
5:688, 2011.
86.	 Omland LH, Jepsen P, Krarup H, et al: Liver cancer and non-Hodgkin
lymphoma in hepatitis C virus-infected patients: Results from the
DANVIR cohort study. Int J Cancer 130:2310, 2012.
87.	 Isikdogan A, Ayyildiz O, Dursun M, et al: Hepatitis C virus in patients
with non-Hodgkin’s lymphoma in southeastern Anatolia region of
Turkey: A prospective case-control study of 119 patients. Leuk Lymphoma
44:1745, 2003.
88.	 Hermine O, Lefrere F, Bronowicki JP, et al: Regression of splenic lym-
phoma with villous lymphocytes after treatment of hepatitis C virus
infection. N Engl J Med 347:89, 2002.
89.	 Awasthi NP, Kashyap R, Kumari N, et al: Sjogren’s syndrome compli-
cated by non-Hodgkin’s lymphoma: Spectrum of lymphoid prolifera-
tions and associated hepatitis C virus infection. Indian J Pathol Microbiol
54:430, 2011.
90.	 Geri G, Terrier B, Semoun O, et al: Surrogate markers of B cell non-
Hodgkin’s lymphoma in patients with hepatitis C virus-related cryo-
globulinaemia vasculitis. Ann Rheum Dis 69:2177, 2010.
91.	 Ennishi D, Maeda Y, Niitsu N, et al: Hepatic toxicity and prognosis in
hepatitis C virus-infected patients with diffuse large B-cell lymphoma
treated with rituximab-containing chemotherapy regimens: A Japanese
multicenter analysis. Blood 116:5119, 2010.
92.	 Ennishi D, Terui Y, Yokoyama M, et al: Monitoring serum hepatitis C
virus (HCV) RNA in patients with HCV-infected CD20-positive B-cell
lymphoma undergoing rituximab combination chemotherapy. Am J
Hematol 83:59, 2008.

Mais conteúdo relacionado

Mais procurados (19)

Lymphoproliferative disorders
Lymphoproliferative disordersLymphoproliferative disorders
Lymphoproliferative disorders
 
Bohomolets Microbiology Lecture #15
Bohomolets Microbiology Lecture #15Bohomolets Microbiology Lecture #15
Bohomolets Microbiology Lecture #15
 
Post Transplant Lymphoproliferative Disorder
Post Transplant Lymphoproliferative DisorderPost Transplant Lymphoproliferative Disorder
Post Transplant Lymphoproliferative Disorder
 
Molecular pathogenesis of head and neck cancer
Molecular pathogenesis of head and neck cancerMolecular pathogenesis of head and neck cancer
Molecular pathogenesis of head and neck cancer
 
Transformation by DNA and RNA viruses
Transformation by DNA and RNA viruses Transformation by DNA and RNA viruses
Transformation by DNA and RNA viruses
 
Oncogenic Viruses
Oncogenic VirusesOncogenic Viruses
Oncogenic Viruses
 
Oncogenesis AAR
Oncogenesis AAROncogenesis AAR
Oncogenesis AAR
 
Ppat.1005652
Ppat.1005652Ppat.1005652
Ppat.1005652
 
Dibakar ppt
Dibakar pptDibakar ppt
Dibakar ppt
 
HPV vaccine (UID198612)
HPV vaccine (UID198612)HPV vaccine (UID198612)
HPV vaccine (UID198612)
 
The Role of Epstein Barr Virsus in Oncogenesis
The Role of Epstein Barr Virsus in OncogenesisThe Role of Epstein Barr Virsus in Oncogenesis
The Role of Epstein Barr Virsus in Oncogenesis
 
Oncogenic viruses
Oncogenic virusesOncogenic viruses
Oncogenic viruses
 
human oncogenic viruses
human oncogenic viruseshuman oncogenic viruses
human oncogenic viruses
 
11 influenzae virus
11 influenzae virus11 influenzae virus
11 influenzae virus
 
Immuno1 overview
Immuno1 overviewImmuno1 overview
Immuno1 overview
 
PNAS-2015-Banerjee
PNAS-2015-BanerjeePNAS-2015-Banerjee
PNAS-2015-Banerjee
 
(20071002)
(20071002)(20071002)
(20071002)
 
Aids
AidsAids
Aids
 
Oncovirus
OncovirusOncovirus
Oncovirus
 

Semelhante a Linfomas y VIH III

Aqueous, Glutinous, Cavitary: Primary Effusion Lymphoma
Aqueous, Glutinous, Cavitary: Primary Effusion LymphomaAqueous, Glutinous, Cavitary: Primary Effusion Lymphoma
Aqueous, Glutinous, Cavitary: Primary Effusion LymphomaCrimsonpublishersTTEH
 
EBV and carcinogenesis.pptx
EBV and carcinogenesis.pptxEBV and carcinogenesis.pptx
EBV and carcinogenesis.pptxssusercd921d1
 
Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Karthick Kumar
 
Cancer and virsues
Cancer and virsuesCancer and virsues
Cancer and virsuesNilesh Kucha
 
JSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.docJSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.docKanchan Parsad
 
journal.pone.0126088.PDF
journal.pone.0126088.PDFjournal.pone.0126088.PDF
journal.pone.0126088.PDFStuart Teal
 
Pathophysiology of lupus nephritis
Pathophysiology of lupus nephritisPathophysiology of lupus nephritis
Pathophysiology of lupus nephritisMohit Mathur
 
Virotherapy,as a key for cancer treatment
Virotherapy,as a key for cancer treatmentVirotherapy,as a key for cancer treatment
Virotherapy,as a key for cancer treatmentShirisha Ratcha
 
Viral carcinogenesis copy
Viral carcinogenesis   copyViral carcinogenesis   copy
Viral carcinogenesis copyAjish Saji
 
molecular basis of lymphomas
molecular basis of lymphomasmolecular basis of lymphomas
molecular basis of lymphomasSamieh Asadian
 
Lymphoproliferative disorders DR MASOUD 2022.pptx
Lymphoproliferative disorders DR MASOUD 2022.pptxLymphoproliferative disorders DR MASOUD 2022.pptx
Lymphoproliferative disorders DR MASOUD 2022.pptxmasoud53
 
A S S I N G M E N T
A S S I N G M E N TA S S I N G M E N T
A S S I N G M E N TZahoor Ahmed
 
5.VIRAL ONCOGENESIS.ppt
5.VIRAL ONCOGENESIS.ppt5.VIRAL ONCOGENESIS.ppt
5.VIRAL ONCOGENESIS.pptINNOCENT66
 

Semelhante a Linfomas y VIH III (20)

Infections and cancer
Infections and cancerInfections and cancer
Infections and cancer
 
Aqueous, Glutinous, Cavitary: Primary Effusion Lymphoma
Aqueous, Glutinous, Cavitary: Primary Effusion LymphomaAqueous, Glutinous, Cavitary: Primary Effusion Lymphoma
Aqueous, Glutinous, Cavitary: Primary Effusion Lymphoma
 
EBV and carcinogenesis.pptx
EBV and carcinogenesis.pptxEBV and carcinogenesis.pptx
EBV and carcinogenesis.pptx
 
Ebv related neoplasms
Ebv related neoplasmsEbv related neoplasms
Ebv related neoplasms
 
Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Epstein barr virus [autosaved]
Epstein barr virus [autosaved]
 
Cancer and virsues
Cancer and virsuesCancer and virsues
Cancer and virsues
 
JSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.docJSResEssay15-Final-TCDkparsad.doc
JSResEssay15-Final-TCDkparsad.doc
 
HIV/AIDS
HIV/AIDSHIV/AIDS
HIV/AIDS
 
journal.pone.0126088.PDF
journal.pone.0126088.PDFjournal.pone.0126088.PDF
journal.pone.0126088.PDF
 
BURKITTS LYMPHOMA
 BURKITTS LYMPHOMA BURKITTS LYMPHOMA
BURKITTS LYMPHOMA
 
Pathophysiology of lupus nephritis
Pathophysiology of lupus nephritisPathophysiology of lupus nephritis
Pathophysiology of lupus nephritis
 
Virotherapy,as a key for cancer treatment
Virotherapy,as a key for cancer treatmentVirotherapy,as a key for cancer treatment
Virotherapy,as a key for cancer treatment
 
Cytokines in diseases
Cytokines in diseasesCytokines in diseases
Cytokines in diseases
 
Viral carcinogenesis copy
Viral carcinogenesis   copyViral carcinogenesis   copy
Viral carcinogenesis copy
 
molecular basis of lymphomas
molecular basis of lymphomasmolecular basis of lymphomas
molecular basis of lymphomas
 
Hepatitis C
Hepatitis C Hepatitis C
Hepatitis C
 
Lymphoproliferative disorders DR MASOUD 2022.pptx
Lymphoproliferative disorders DR MASOUD 2022.pptxLymphoproliferative disorders DR MASOUD 2022.pptx
Lymphoproliferative disorders DR MASOUD 2022.pptx
 
CAR T CELL
CAR T CELLCAR T CELL
CAR T CELL
 
A S S I N G M E N T
A S S I N G M E N TA S S I N G M E N T
A S S I N G M E N T
 
5.VIRAL ONCOGENESIS.ppt
5.VIRAL ONCOGENESIS.ppt5.VIRAL ONCOGENESIS.ppt
5.VIRAL ONCOGENESIS.ppt
 

Mais de Marcela Alejandra Casas (16)

Linfomas y VIH II
Linfomas y VIH IILinfomas y VIH II
Linfomas y VIH II
 
Linfomas y VIH I
Linfomas y VIH ILinfomas y VIH I
Linfomas y VIH I
 
Linfomas y VIH, a proposito de un caso...
Linfomas y VIH, a proposito de un caso...Linfomas y VIH, a proposito de un caso...
Linfomas y VIH, a proposito de un caso...
 
INPPB
INPPBINPPB
INPPB
 
Breve resumen IPPB
Breve resumen IPPBBreve resumen IPPB
Breve resumen IPPB
 
Osteo3
Osteo3Osteo3
Osteo3
 
Biofilms
BiofilmsBiofilms
Biofilms
 
Infecciones asociadas a protesis
Infecciones asociadas a protesisInfecciones asociadas a protesis
Infecciones asociadas a protesis
 
SAMR antiguo no obstante util...
SAMR antiguo no obstante util...SAMR antiguo no obstante util...
SAMR antiguo no obstante util...
 
MRSA
MRSAMRSA
MRSA
 
SIRI
SIRISIRI
SIRI
 
SIRI
SIRISIRI
SIRI
 
Meningoencefalitis TB
Meningoencefalitis TBMeningoencefalitis TB
Meningoencefalitis TB
 
CMV
CMVCMV
CMV
 
Resistencia ATB 2014
Resistencia ATB 2014Resistencia ATB 2014
Resistencia ATB 2014
 
Antigpp65
Antigpp65Antigpp65
Antigpp65
 

Último

Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...aartirawatdelhi
 
Top Rated Bangalore Call Girls Richmond Circle ⟟ 9332606886 ⟟ Call Me For Ge...
Top Rated Bangalore Call Girls Richmond Circle ⟟  9332606886 ⟟ Call Me For Ge...Top Rated Bangalore Call Girls Richmond Circle ⟟  9332606886 ⟟ Call Me For Ge...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 9332606886 ⟟ Call Me For Ge...narwatsonia7
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...narwatsonia7
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...Arohi Goyal
 
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...perfect solution
 
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiRussian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiAlinaDevecerski
 
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...vidya singh
 
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...indiancallgirl4rent
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...narwatsonia7
 

Último (20)

Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
 
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
 
Top Rated Bangalore Call Girls Richmond Circle ⟟ 9332606886 ⟟ Call Me For Ge...
Top Rated Bangalore Call Girls Richmond Circle ⟟  9332606886 ⟟ Call Me For Ge...Top Rated Bangalore Call Girls Richmond Circle ⟟  9332606886 ⟟ Call Me For Ge...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 9332606886 ⟟ Call Me For Ge...
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
 
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
 
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 9907093804 Top Class Call Girl Service Available
 
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
 
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
 
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiRussian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
 
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
 
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Siliguri Just Call 9907093804 Top Class Call Girl Service Available
 
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
(Rocky) Jaipur Call Girl - 09521753030 Escorts Service 50% Off with Cash ON D...
 
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...Bangalore Call Girls Nelamangala Number 7001035870  Meetin With Bangalore Esc...
Bangalore Call Girls Nelamangala Number 7001035870 Meetin With Bangalore Esc...
 

Linfomas y VIH III

  • 1. 1245 C H A P T E R 82 VIRUS-ASSOCIATED LYMPHOMA Jennifer A. Kanakry and Richard F. Ambinder There are five well-characterized human viruses that are generally accepted as important in lymphomagenesis (Table 82-1). These viruses may infect tumor cells (or their progenitors) or may act at a distance. The genomes of Epstein-Barr virus (EBV), Kaposi sarcoma– associated herpesvirus (KSHV, also known as human herpesvirus 8 [HHV-8]), and human T-lymphotropic virus-1 (HTLV-1) are present in tumor cells. The viral genes expressed in tumor cells modulate cellular metabolism, proliferation, and cell death. In contrast, the human immunodeficiency virus (HIV) genome is generally not detected in tumor cells. Whether hepatitis C virus (HCV) genomes are present in lymphoma cells remains a subject of controversy. Although viral infection plays a role in the pathogenesis of some lymphomas, lymphomagenesis is unusual. Only a small subset of infected people develops lymphoma. Furthermore, although primary viral infection may be followed by lymphomagenesis within days or weeks in exceptional circumstances, most lymphomas arise years or decades after primary infection. Indeed the term adult in adult T-cell leukemia/lymphoma (ATL) reflects the time lag between HTLV-1 infection in infancy and the evolution to malignancy. Geography and associated environmental exposures, host genetic factors, and immune status all modify risk. Aspects of the biology and epidemiology of each of these viruses and their relationship with lymphomagenesis are reviewed. In addi- tion, clinically important and distinctive features of diagnosis and treatment of the associated lymphomas are presented. EPSTEIN-BARR VIRUS Viral Biology EBV is a gammaherpesvirus transmitted mainly through saliva.1,2 After primary infection, some of the infected cells are driven to pro- liferate and thereby spread infection throughout the B-cell compart- ment. Ultimately, in the normal host, there is an immune response that controls infection and eradicates virus-infected proliferating cells. Thereafter the viral genome is harbored mainly in resting memory B lymphocytes that persist for life. These B cells that harbor virus elude immune surveillance in part because of their very restricted viral gene expression such that few viral antigens are presented. Occa- sionally there is activation of viral lytic gene expression (at least in some instances this occurs in concert with plasma cell differentiation) leading to production of infectious virions that may infect other B cells. T cell–mediated immune function keeps such proliferation in check.3 In vitro EBV immortalizes B cells such that they grow indefinitely as lymphoblastoid cell lines (LCLs) (Fig. 82-1). LCLs are tumorigenic in immunodeficient mice. In LCL, viral genomes are present as cir- cular double-stranded deoxyribonucleic acid (DNA) episomes within the nucleus. The viral proteins required for immortalization include Epstein-Barr virus nuclear antigen-1 (EBNA1), a sequence-specific DNA-binding protein important in the maintenance of the viral episome; EBNA2, a transcription factor that has many effects similar to those of activated Notch receptors; and latent membrane protein-1 (LMP1), a constitutively activated member of the tumor necrosis factor (TNF) receptor superfamily, which most closely resembles CD40.4 LMP1 activates the nuclear factor kappa-B (NFκB) pathway, which modulates cell proliferation and apoptosis.5 Several other EBV proteins are also required for immortalization. Although EBV immor- talization of B cells in vitro may offer some insights into tumorigen- esis, some caution is required in using LCL as a tumor model. Most EBV tumors, including tumors of B-lineage cells, do not express many of the viral genes required for lymphocyte immortalization. The only tumors that express the full complement of viral proteins required for immortalization are those that arise in the most pro- foundly immunocompromised patients (organ or hematopoietic transplant recipients, patients with congenital immunodeficiency, or patients with far advanced acquired immunodeficiency syndrome [AIDS]). Thus in posttransplantation lymphoproliferative disorder (PTLD), tumor cells may resemble LCL in expressing many viral latency genes in association with normal karyotype (and few muta- tions of the cellular genome). It has been suggested that there is an inverse relationship between cellular mutations and viral gene expres- sion in tumors.6 EBV gene expression may directly drive proliferation or inhibit apoptotic pathways as illustrated by lymphocyte immortalization. However, viral gene expression may also perturb normal lymphocyte biology. Thus LMP1 expression upregulates activation induced (cyti- dine) deaminase expression, which facilitates somatic hypermutation and immunoglobulin class switching.7 LMP1 expression may also be important in the conversion of naive B-cells to post-germinal center memory B-cells. LMP2A allows B-cells that lack normal immuno- globulin expression to escape regulatory checkpoints and survive.8 Epidemiology of Viral Infection EBV infection is ubiquitous. The vast majority of adults are infected worldwide. Primary infection is most often asymptomatic, especially when it occurs in childhood.9 Primary infection may be associated with the syndrome of infectious mononucleosis. Symptomatic primary infection occurs more frequently in older children and in adults than in younger children. Other possible determinants of symptomatic primary infection include genetic factors and possibly the size of the viral inoculum. Strain differences in EBV are well recognized.10 However, the importance of these strain differences with regard to lymphomagen- esis remains poorly understood. There is general agreement that the Type 1 strain EBV is most common worldwide and in tumors. The Type 2 strain virus has been identified in some African Burkitt lym- phoma (BL) and in some AIDS-associated lymphoma. A-strain virus is more efficient at lymphocyte immortalization in vitro and lym- phomagenesis in mouse models. The two strains of virus differ mainly in the EBNA2 gene, but differences are recognized in some other viral proteins as well.11 Variations in the regulatory regions or coding regions of a variety of other genes including EBNA1, LMP1, and ZTA have been recognized and suggested to play a role in lymphomagenesis. A simple classification of latent viral gene expression recognizes three patterns as shown in the Table 82-2.
  • 2. Part VII  Hematologic Malignancies1246 at very high copy number (perhaps millions of copies per cell) in latently infected cells. The function(s) of these RNAs is disputed, but their use for the detection of virus in a variety of surgical specimens is generally accepted. Viral antigens are detected by immunohistochemistry. In clinical laboratories,immunohistochemistryforLMP1iscommonlyemployed and is sensitive for the detection of EBV in Hodgkin lymphoma (HL). In a variety of other EBV-associated B- andT-cell malignancies, expres- sion is variable. Thus failure to detect LMP1 expression does not exclude the presence of EBV except perhaps in HL. In principle, detec- tion of EBNA1 should be universally applicable, although the low level of antigen expression and the cross-reactivity of available monoclonal antibodies have prevented immunohistochemistry for this antigen from emerging as a standard tool. Association With Particular Types of Lymphoma Some lymphoma types are nearly 100% EBV associated, including endemic BL, extranodal natural killer (NK)/T-cell lymphoma of the nasal type, early PTLD, lymphomatoid granulomatosis, diffuse large B-cell lymphoma (DLBCL) associated with chronic inflammation, EBV-positive DLBCL of older adults, and AIDS primary central nervous system (CNS) lymphoma (PCNSL).13-16 Other lymphoma types are variably EBV associated. These include classic HL, PTLDs occurring many months or years after transplantation, and systemic AIDS-related lymphoma. Some lymphoma types appear never or almost never to be EBV associated, including follicular lymphoma, nodular lymphocyte-predominant HL, and mantle cell lymphoma. Table 82-3 lists the lymphomas that have been associated with EBV, associated cofactors, viral antigen expression, and an estimate of the percentage of tumors within each lymphoma subtype that harbor viral genomes. Posttransplantation Lymphoproliferative Disorder PTLD is a group of lymphoproliferative disorders ranging from poly- clonal lymphoid hyperplasia to lymphomas that arise in patients after solid organ or hematopoietic stem cell transplantation (HSCT).17 PTLD, especially in the first few months after transplantation, is highly associated with EBV (Fig. 82-2, A, B). EBV gene expression in PTLD corresponds to latencies 2 and 3.18 Broad expression of viral proteins is seen only in immunosuppressed hosts, perhaps reflecting that many of these proteins are commonly targeted by cytotoxic T cells. B cells that harbor EBV are able to proliferate in the setting of posttransplantation immunosuppression at least in part due to decreased T-cell surveillance.19 HSCT patients that receive grafts that have been T-cell depleted develop EBV-associated PTLD at very high rates. Treatment of rejection in solid organ transplant recipients with agents such as the monoclonal antibody OKT3, which targets CD3+ cells, is associated with markedly increased risk for PTLD.20 Treat- ment strategies such as the use of rituximab and infusion of EBV- specific cytotoxic T cells have been quite effective in treating or preventing PTLD (see box on Epstein-Barr Virus-Associated Positive Posttransplant Lymphoproliferative Disorder). Figure 82-1  EPSTEIN-BARR VIRUS (EBV)–IMMORTALIZED B CELL. Normal B cells are readily immortalized in vitro with EBV. These cells express EBV nuclear and membrane proteins. The nuclear proteins include a protein expressed in all EBV-associated tumors, Epstein-Barr virus nuclear antigen 1 (EBNA1). This protein is required for episomal maintenance. Other viral nuclear proteins expressed are transcription factors. These include EBNA-LP, EBNA2, EBNA3A, EBNA3B, and EBNA3C. Two membrane proteins are expressed: latent membrane protein 1 (LMP1), which activates NFκB path- ways, and LMP2A, which mimics B-cell receptor (immunoglobulin) signaling. LMP2 LMP1 NF-κB Cell survival B-cell receptor EBNA1 Transcription factors: EBNA-LP EBNA2 EBNA3A, EBNA3B, EBNA3C Table 82-1  Viruses and Lymphomagenesis Virus Viral Genome in Tumor Cell Lymphoma Type EBV Episomal B, T, NK KSHV Episomal B HTLV-1 Integrated T HIV-1 Absent B HCV Uncertain B EBV, Epstein-Barr virus; HCV, hepatitis C virus; HIV-1, human immunodeficiency virus type 1; HTLV-1, human T-lymphotropic virus-1; KSHV, Kaposi sarcoma–associated herpesvirus; NK, natural killer. Table 82-2  Patterns of Epstein-Barr Virus Gene Expression in Latency Latency EBNA1 EBNA2, EBNA3A, EBNA3B, EBNA3C LMP1 LMP2A I + II + + + III + + + + EBNA1, Epstein-Barr virus nuclear antigen 1; EBV, Epstein-Barr virus; LMP1, latent membrane protein 1. Epstein-Barr Virus Detection in Clinical Specimens The sensitivity of polymerase chain reaction (PCR) makes detection of viral DNA straightforward, but the ubiquity of EBV infection, and the persistence of B cells that harbor EBV in all seropositive individu- als, means that EBV DNA is readily detected in many specimens that include normal lymphocytes. Thus diagnosis of the EBV association in general requires viral detection specifically in tumor cells. Tech- niques for viral DNA detection by fluorescence in situ hybridization or related techniques, although greatly improved in recent years, remain the purview of research laboratories and are generally not readily applicable to clinical specimens. This reflects the relatively low copy number of the viral genome in tumor cells, typically 1 to 200 copies per cell. In contrast, in situ hybridization for the Epstein-Barr encoded ribonucleic acids (RNAs) has emerged as a laboratory stan- dard.12 These RNAs are polymerase 3 transcripts that are expressed
  • 3. Chapter 82  Virus-Associated Lymphoma 1247 Hodgkin Lymphoma Approximately 30% of HL tumors in the United States and Europe are EBV associated (Fig. 82-2, C, D).21,22 Epidemiologic studies in Denmark and Sweden suggest that individuals with a history of symptomatic infectious mononucleosis are at increased risk for EBV- associated HL, but not for EBV-negative HL or other lymphomas.23 The period of risk peaks at about 2 years but continues to be elevated for at least 10 years after symptomatic mononucleosis. Figure 82-2  EXAMPLES OF EPSTEIN-BARR VIRUS (EBV)–RELATED LYMPHOMAS. Posttransplantation lym- phoproliferative disorder (PTLD), polymorphic type, EBV+ occurring in the gastrointestinal tract of a 15-month-old girl following an orthotopic liver transplant (A and B). There was a mildly atypical lymphocytic infiltrate in the duodenal mucosa composed of small lymphocytes, plasma cells, and occasional large cells (A). The infiltrate was EBV+ as dem- onstrated by in situ hybridization for Epstein-Barr–encoded RNA (EBER) (B). Hodgkin lymphoma, nodular sclerosing type, EBV+ (C and D). The hematoxylin-eosin section shows a portion of a lymph node from a cervical lymph node biopsy of an 8-year-old girl. There are bands of sclerosis forming a cellular nodule, and within the nodule there is a mixed inflammatory infiltrate and scattered large cells with contracted cytoplasm consistent with lacunar cells (C). The immunophenotype of these cells was that of classic Hodgkin lymphoma, and the cells were EBER+ (D). EBV is seen more frequently in mixed cellularity Hodgkin lymphoma but can be seen in 10% to 40% of cases of nodular sclerosing type. It is even more frequent in cases associated with human immunodeficiency virus (HIV; see text) and in resource- poor regions. Burkitt lymphoma, sporadic type, EBV+ (E and F). A section of the cervical lymph node biopsy of a 9-year-old girl with a rapidly enlarging neck mass is shown. The section illustrates the classic morphologic features of Burkitt lymphoma with a “starry sky” appearance, sheets of intermediate-sized cells with multiple small nucleoli and high mitotic rate. The cells were virtually all EBER+ (F). EBV can be seen in about 20% to 30% of cases of sporadic Burkitt lymphoma and is essentially always positive in endemic cases. F A C E B D F A E Table 82-3  Epstein-Barr Virus-Associated Lymphoma Type Cofactors Viral Gene Expression Approximate % EBV Associated Comment PTLD Immunosuppression, allograft Latency II or III 50-95 Early days/months after transplantation are more commonly associated with EBV Sporadic BL Latency I 20 in the United States Higher in Latin America Endemic BL Malaria Latency I >95 AIDS BL HIV Latency I 30 HL Latency II 30 in the United States Higher % in mixed cellularity, in males, in Hispanics AIDS PCNSL HIV Latency II or III >95 Nasal-type NK cell More common in Asia Latency II >95 AIDS PEL HIV and KSHV Latency I >75 AIDS, Acquired immunodeficiency syndrome; BL, Burkitt lymphoma; EBV, Epstein-Barr virus; HIV, human immunodeficiency virus; HL, Hodgkin lymphoma; KSHV, Kaposi sarcoma–associated herpesvirus; NK, natural killer; PCNSL, primary central nervous system lymphoma; PEL, primary effusion lymphoma; PTLD, posttransplantation lymphoproliferative disorder. Higher EBV associations are seen in Latin America, Africa, and parts of Asia. Factors associated with EBV tumor positivity include mixed cellularity and lymphocyte-depleted classic HL histo- logic subtypes, male gender, low socioeconomic background, history of symptomatic infectious mononucleosis, and Hispanic ethnic- ity.21,24,25 Organ and hematopoietic stem cell transplant recipients and HIV-positive patients are more likely to develop HL than the general population, and approximately 90% of the tumors are EBV associated.26
  • 4. Part VII  Hematologic Malignancies1248 Figure 82-3  EXAMPLES OF KSHV- AND HTLV-1-ASSOCIATED LYMPHOPROLIFERATIVE DISEASE. Primary effusion lymphoma (A and B). A, The pleural tap had a high cell count, and the cytospin preparation revealed a markedly pleomorphic cell population with large and giant cells with deep-blue cytoplasm. B, A cell block was pre- pared (top) so that in situ hybridization studies could be performed. These studies showed the cells to be KSHV+ by immunohistochemistry for latency associated nuclear antigen-1 (LANA-1) (bottom) and EBV+ by EBER in situ hybrid- ization (not shown). C, Adult T-cell leukemia/lymphoma in a patient who was HTLV-1+. The peripheral smear showed the classic “flower” cells. EBER, Epstein-Barr–encoded RNA; HTLV-1, human T-lymphotropic virus-1; KSHV, Kaposi sarcoma–associated herpesvirus. (A and B courtesy Dr. Elizabeth Hyjek, University of Chicago.) A B C Epstein-Barr Virus-Associated Positive Posttransplant Lymphoproliferative Disorder A 55-year-old renal transplant patient presents with acute renal failure 5 months after transplant. She is found on imaging to have an obstructing mass in the transplanted kidney. She under- goes kidney biopsy, and Epstein-Barr virus (EBV)–positive post- transplant lymphoproliferative disorder (PTLD) involving the transplanted organ is diagnosed. Treatment options include ritux- imab; decreasing immunosuppression (acknowledging the associ- ated risk for organ rejection); changing immunosuppressive agents—switching a calcineurin inhibitor for a mammalian target of rapamycin (mTOR) inhibitor; combination chemotherapy; or, in the case of renal transplant, removal of the transplanted organ and withdrawal of immunosuppression. The EBV gene expression pattern in HL is latency II even when HL occurs in immunocompromised populations.27 LMP1 and LMP2A may mimic signaling of B-cell receptors and thus protect B cells lacking functional immunoglobulin expression from apoptotic signaling. Approximately 20% of HL lack productive immunoglobu- lin gene rearrangements. These tumors appear to be exclusively EBV associated. The EBV association of HL appears not to have prognostic import in young adult patients but is associated with poorer survival in older patients in several reports.22 Treatment strategies involving adoptive therapy with EBV-specific T cells expanded in vitro are under study.28 Burkitt Lymphoma Endemic BL is nearly 100% associated with EBV, whereas sporadic and HIV-associated BL are much more variably EBV associated (see Table 82-3 and Fig. 82-2, E, F).29 Viral expression is latency I (i.e., EBNA1 is the only viral protein consistently expressed). The defining feature of BL is a translocation between c-Myc on chromosome 8 and one of the immunoglobulin genes on chromosomes 2, 14, or 22. It has been generally presumed that falciparum malaria is a cofactor in endemic BL, and the distribution of BL in Africa corresponds to the distribution of holoendemic malaria. However, little is understood of the pathogenesis or interaction between these infectious cofactors. The characteristic presentation of BL is different in the endemic versus sporadic settings, but there is no evidence to link these presentations specifically with the virus. The virus-tumor association does not guide diagnosis, therapy, or estimation of prognosis at present. Diffuse Large B-cell Lymphoma Associated With Chronic Inflammation EBV-associated lymphomas sometimes arise in the setting of long- standing chronic inflammation.15 This was first described in Japanese patients with a remote history of pulmonary tuberculosis treated with thoracoplasty with resulting chronic pyothorax, although many more cases have since been reported. These patients developed EBV- associated DLBCL of the pleural lining and associated lung tissue decades after thoracoplasty. Similar cases of aggressive, EBV-associated B-cell lymphomas have been reported to arise in patients at the sites of chronic inflammation associated with various implants, surgical mesh, or in the lung after chronic empyema. KAPOSI SARCOMA–ASSOCIATED HERPESVIRUS Virus and Tumor Epidemiology KSHV (HHV-8) is a gammaherpesvirus that, unlike EBV, has a low prevalence worldwide.30 The virus is endemic in certain areas, such as in sub-Saharan Africa and the Middle East, and has an intermedi- ate prevalence in Mediterranean countries. Transmission is believed to be predominately through saliva. Similar to EBV, KSHV latently infects B cells; viral genes that promote cell survival are implicated in lymphomagenesis. KSHV was discovered in Kaposi sarcoma but is also present in two lymphoproliferative diseases: primary effusion lymphoma (PEL) (Fig. 82-3, A, B) and multicentric Castleman disease (MCD).16,31 PEL occurs almost exclusively in HIV-positive patients, particularly in men who have sex with men, and typically when CD4 counts are less than 100/mm3 . PELs are usually dually infected with KSHV and EBV, but KSHV is always present. KSHV-associated MCD, although much more common in HIV-infected populations, also occurs in the general population. MCD is a KSHV-associated, immunoglobulin M (IgM) λ– producing nonclonal lymphoproliferative disorder typically involving the mantle zone of lymph nodes and the spleen. The KSHV-positive cells in MCD are always EBV negative. These cells express a broad range of KSHV lytic antigens, and high KSHV copy numbers are reported in plasma. Evolution into or coassociation with an aggressive lymphoma, often of plasmablastic phenotype, is not uncommon.
  • 5. Chapter 82  Virus-Associated Lymphoma 1249 HUMAN T-LYMPHOTROPIC VIRUS-1 Viral Biology HTLV-1 is a retrovirus with a single-stranded RNA genome.37,38 Fol- lowing infection there is reverse transcription and integration of proviral DNA into the host genome. HTLV-1 infects a variety of cell types but persists in CD4+ T lymphocytes. Viral infection within the host is spread from cell to cell through direct cell-to-cell contact. As with EBV, the proliferation of HTLV-1–infected lymphocytes plays a central role in ensuring viral persistence. The HTLV-1 protein Tax plays a central role in T-lymphocyte immortalization and transformation (Fig. 82-4).37,39 Tax affects NFκB and the serine/threonine kinase AKT pathways with diverse prolifera- tive and antiapoptotic effects. As with some of the immunodominant EBV antigens expressed in proliferating lymphocytes, Tax expression is targeted by cytotoxic T cells. Another viral protein, Hbz, suppresses Tax expression, allowing transformed T cells to elude immune sur- veillance.40 Tax interferes with various DNA repair pathways and induces reactive oxygen species, facilitating the development of aneu- ploidy.37,39 Tax leads to functional inactivation of p53 and may inter- fere with the spindle assembly checkpoint that normally operates in mitosis to preserve euploidy. Epidemiology of Viral Infection and Adult T-Cell Leukemia/Lymphoma HTLV-1 is endemic in particular regions of Japan, Africa, South America, and some Caribbean islands.40,41 As assessed by seropreva- lence, rates up to 37% are found on the southwestern Japanese islands of Shikoku, Kyushu, and Okinawa, whereas most other areas of Japan have an intermediate prevalence of 1% to 5%. In the United States Within the HIV population, nearly all cases of MCD are KSHV associated; this viral association is not as strong among HIV-negative MCD patients. High expression of viral interleukin-6 (vIL-6) is thought to contribute to the systemic inflammation seen in this disorder.32 The KSHV-associated lymphomas that arise in association with MCD are not nodal equivalents of PEL; rather these plasma- blastic lymphomas are uniformly EBV negative and express IgM λ.16 Diagnostic and Therapeutic Considerations The requisite finding in PEL is a lymphomatous effusion, which can be pleural, pericardial, or peritoneal, without associated lymphade- nopathy or masses, arising in the setting of immunocompromise. Often, patients with HIV/AIDS will present with PEL in addition to other KSHV-associated diseases, such as Kaposi sarcoma and MCD, so thorough evaluation and staging should be undertaken at diagnosis. On cytologic examination, the PEL tumor cells are large with prominent nucleoli. The effusion cells are clonal B cells with CD45 positivity but typically lack other specific B-cell lineage markers, although CD30 and CD38 positivity can be seen. BCL6 mutations are frequently detected. Tumor cells always harbor KSHV as demonstrated by staining for KSHV-associated latency-associated nuclear antigen-1 (LANA-1). In MCD there is characteristically λ light chain restriction.33 This does not reflect clonality as assessed by study of Ig DNA rearrangements. Rather it reflects a tendency for the virus to selectively infect cells expressing λ or to selectively drive such cells to proliferate. Treatment of PEL is most commonly combination chemotherapy. Outcomes remain quite poor. Treatment of MCD with targeted therapies has been more successful, including the use of rituximab and IL-6 inhibition.34,35 Antivirals such as ganciclovir and valganci- clovir have shown clinical activity in MCD, because viral lytic replica- tion is a feature of this disease.36 Figure 82-4  HTLV-1 AND THE EVOLUTION OF ADULT T-CELL LEUKEMIA/LYMPHOMA. Following HTLV-1 infection, many cells undergo apoptosis, but some infected CD4+ T cells are driven to proliferate by the effects of Tax on the nuclear factor kappa-B (NFκB) pathway and on the AKT pathway. Tax is also suggested to result in inactivation of p53, aneuploidy, and deoxyribonucleic acid (DNA) damage. Over many decades malignancy evolves. HTLV-1, Human T-lymphotropic virus-1. HTLV-1 infection Tax activates AKT and NF-κB, inactivates p53 Apoptosis Tax Tax Tax Tax Viral control by cytotoxic T lymphocytes CTL Proliferation Tax induces aneuploidy and DNA damage 30-40 years Cellular transformation to ATL
  • 6. Part VII  Hematologic Malignancies1250 infection may lead to cell death or establishment of latency in resting cells. HIV infection is spread either through new rounds of virion production with cellular infection or cell-cell fusion. There is no evidence to suggest that infected cells are driven to proliferate. This is in contrast to HTLV-1 and EBV, where proliferation of infected cells appears to play a key role in establishing the long-term viral reservoir and perhaps in mediating lymphomagenesis. The lympho- mas that are increased in HIV-infected patients (Table 82-4) are of B-cell lineage, and there is no substantial evidence that HIV infection of B cells is important in the pathogenesis of these lymphomas. Rather it appears that the HIV infection compromises cellular immu- nity, decreasing immune surveillance of EBV- and KSHV-infected B cells. In addition, HIV infection stimulates proliferation of B lym- phocytes and perhaps genetic aberrations in the proliferating cells.51 Many possible mechanisms have been invoked, including (1) direct stimulation of B cells by HIV antigens or antigens associated with opportunistic infection; (2) stimulation of B cells by cellular proteins (CD40 ligand) incorporated into the HIV virion leading to expres- sion of activation-induced deaminase, an enzyme that mediates double-strand DNA breaks; and (3) dysregulation of B cells as a consequence of T-cell dysfunction.16,51,52 There is also some evidence that host biology may contribute to lymphomagenesis. In particular, there are a variety of genetic poly- morphisms that influence susceptibility to HIV-1 infection, such as CCR5-Δ32.53,54 Individuals who are homozygous for this polymor- phism are much less likely to be infected by HIV than others. Some evidence has emerged to suggest that in heterozygotes who are infected, HIV progression is slowed and there is less likelihood of lymphomagenesis. Many of these lymphomas are associated with EBV, KSHV, or both (see Table 82-4). Epidemiology Lymphoma is increased in all HIV risk groups, in contrast to Kaposi sarcoma, which is rarely seen in injection drug users (or in hemo- philiacs in an earlier era).55 There is a well-established relationship between CD4+ cells per cubic millimeter overall and the risk for lymphoma, but the relationship is complex and differs among lym- phomas (see Table 82-4). The incidence of non-Hodgkin lymphoma (NHL) in the HIV population, particularly PCNSL (Fig. 82-5, A to C), has decreased with the widespread use of highly active antiretroviral therapy (HAART), although HIV patients on HAART still carry an increased risk for lymphoma compared to the HIV-negative population.55 In the HAART era, HIV patients on average have higher CD4+ counts (often greater than 100 cells/mm3 ) when diagnosed with lymphoma as compared to the pre-HAART era. Among HIV patients with very low CD4+ counts,56 NHLs are still seen at rates similar to the pre- HAART era. Patients with HIV are now living much longer because of effective antiretroviral regimens and decreased rates of opportunis- the incidence in blood donors is 0.025%. It has been estimated that about 20 million individuals are infected worldwide. The major mode of transmission in endemic areas is from mother to child in breast milk, although infection is also transmitted through sexual intercourse, transfusion of cellular blood products, and the sharing of needles and syringes. Evidence has been presented that HTLV-1 infection persists in association with certain human leukocyte antigen (HLA) types and may be more readily transmitted from mother to child when these HLA types are shared. ATL is more common in men than in women and typically pres- ents in the fourth or fifth decade of life. Perhaps as a function of the long latency period, cases of ATL following blood transfusion or needle sharing are vanishingly rare. The lifetime risk for ATL has been estimated to be in excess of 6% in men who are HTLV-1 carriers in an endemic region of Japan, although in other settings the risk may be much lower.42 As with EBV, the subset of the infected population that develops lymphoma is quite small. Adult T-Cell Leukemia/Lymphoma Diagnostic Considerations There is a spectrum of HTLV-1–associated leukemia/lymphoma.38 Twenty percent of ATL cases present with a lymphomatous subtype dominated by lymphadenopathy and hepatosplenomegaly. Cutane- ous infiltration, lytic bone lesions, malignant effusions, and involve- ment of the CNS and other extranodal sites are not uncommon.40 Hypercalcemia is common, particularly in leukemic forms. Presenta- tions reflecting immune dysfunction such as strongyloidiasis with dissemination, Pneumocystis jiroveci pneumonia (PJP), mycobacte- rium, or cryptococcal infection are common. In the leukemic subtype of ATL, the classic findings on peripheral blood smear are lympho- cytes with flower-shaped nuclei (Fig. 82-3, C). Histologically ATL shows lymph node effacement by large, atypi- cal T cells usually expressing CD4+ , CD25+ , and CD52+ , with vari- able CD30 and CD15 expression. Aneuploidy is consistent, although characteristic cytogenetic abnormalities have not been identified. Serologic analysis confirms the presence of HTLV-1 infection.38 Therapies Specific to HTLV-1 ATL ATL is an aggressive lymphoma with poor responses to chemother- apy, high relapse rates, and low overall survival. More indolent chronic and smoldering forms are also recognized. Combination chemotherapy regimens used in the treatment of other lymphomas or leukemias are commonly used, but no standard regimen has emerged.38,43 Several antivirals used in the treatment of HIV infection have activity against HTLV-1. Among them are zidovudine and lami- vudine. The combination of interferon and zidovudine has yielded promising results, particularly in the leukemic subtype.38 Proteasome and histone deacetylase (HDAC) inhibitors have attracted interest. Given high CD25 and CD52 expression in ATL, the efficacy of monoclonal antibodies aimed against these two receptors has been investigated. Thus far alemtuzumab, an anti-CD52 monoclonal anti- body, may have some activity in ATL based on small phase II studies and case reports.44,45 Arsenic trioxide combined with interferon-α has been shown to induce remissions in relapsed, refractory patients with ATL, although durable responses were limited. In a trial of 20 ATL patients, all-trans retinoic acid was used with 40% achieving remis- sion. Allogeneic hematopoietic transplantation has been increasingly recognized as an effective therapy for ATL.46,47 HIV-ASSOCIATED LYMPHOMAS Viral Biology and Pathogenesis HIV-1 is a retrovirus that infects CD4+ T cells and monocytes.48,49 It appears to establish a lifelong reservoir in CD4+ T cells.50 Viral Table 82-4  HIV-Associated Lymphoma Lymphoma CD4 Association (cells/mm3 ) EBV Association (%) Other Cofactors PCNSL <50 >95 DLBCL Variable 40 BL >100 30 HL >100 90 PEL <100 >75 KSHV BL, Burkitt lymphoma; DLBCL, diffuse large B-cell lymphoma; EBV, Epstein- Barr virus; HIV, human immunodeficiency virus; HL, Hodgkin lymphoma; KSHV, Kaposi sarcoma–associated herpesvirus; PCNSL, primary central nervous system lymphoma; PEL, primary effusion lymphoma.
  • 7. Chapter 82  Virus-Associated Lymphoma 1251 Diagnostic Considerations Specific to Lymphoma in Patients With HIV Lymphomas in HIV-infected patients are more likely to present with B symptoms such as fever and night sweats, and advanced stage, including bone marrow, extranodal, and CNS involvement. Thus the approach to diagnosis is somewhat different than the approach in the HIV-negative patient. Unexplained fever and sweats in an HIV-seropositive patient, even in the absence of lymphadenopathy, are sufficient to warrant consid- eration of HL. Patterns of disease involvement also differ in HIV- infected patients. Contiguous spread so characteristic of classic HL in other settings is less common in HIV HL. Bone marrow–only presentations of HL are not uncommon (Fig. 82-5, D to G), and the possibility of HL should be in the differential diagnosis even in the absence of lymphadenopathy. Patients with HIV-associated NHL have higher rates of extranodal involvement, including bone marrow and CNS disease, as well as higher stage disease and more aggressive tumors on average.72 There- fore it is recommended that all HIV-seropositive patients with aggres- sive NHL undergo a diagnostic lumbar puncture. The routine use of CNS intrathecal chemotherapy prophylaxis in all HIV-seropositive NHL patients is controversial but reasonable and typically done in particularly high-risk patients, such as those with BL, marrow or testicular involvement, or extranodal disease. Imaging is often more difficult to interpret in HIV patients than in other settings. Lymphadenopathy associated with HIV infection or opportunistic infection is common, and the presumption that enlarged lymph nodes reflect the presence of lymphoma in patients with known lymphoma or history of lymphoma is not as safe as in tic infection. As a result, malignancy has emerged as the major cause of mortality in HIV populations with access to antiretroviral therapy.57,58 With the widespread use of HAART, the incidence of HL in HIV- seropositive patients has not declined; patients with CD4+ counts between 150 and 199 cells/mm3 actually have higher risk for HIV-associated HL than patients with CD4+ counts of less than 50 cells/mm3 .59 HIV Hodgkin Lymphoma A 42-year-old human immunodeficiency virus (HIV)–positive patient, on highly active antiretroviral therapy (HAART) with a CD4 count of 324 cells/mm3 and undetectable viral load, pres- ents with fever, weight loss, and a palpable axillary lymph node. Hodgkin lymphoma (HL) is diagnosed on excisional biopsy, and Epstein-Barr virus (EBV) positivity is demonstrated by Epstein- Barr–encoded RNA (EBER) in the Reed-Sternberg tumor cells. Although he lacks additional lymphadenopathy, a bone marrow biopsy is performed, and it shows involvement by HL. He has stage IVB disease. Pneumocystis jiroveci pneumonia prophylaxis is started despite adequate CD4 count in anticipation of chemo- therapy. His HAART regimen is reviewed for potential antiviral- chemotherapy drug interactions. He receives six cycles of full-dose, first-line chemotherapy and achieves a complete remission. Figure 82-5  EXAMPLES OF HUMAN IMMUNODEFICIENCY VIRUS (HIV)–RELATED LYMPHOMAS. Primary central nervous system (CNS) lymphoma in HIV+ patients (A to C). Gross appearance (coronal section) of the brain from an autopsy of a 24-year-old HIV+ female patient with temporo- parietal mass due to a primary CNS lymphoma (A). The patient died from uncal and cingulate herniation. Biopsy section from another patient showing a perivascular infiltrate of large lymphoma cells. This is the typical pattern of involvement by CNS lymphoma (B). The cells were shown to be CD20+ B cells and were EBER+ (C). A diagnosis can be made without biopsy when magnetic resonance imaging studies show characteristic features and EBV is demonstrated in the cerebrospinal fluid by polymerase chain reaction (see box on AIDS Primary Central Nervous System Lymphoma). Hodgkin lymphoma extensively involving the bone marrow in an HIV+ patient with stage IVB disease (D to G). The bone marrow biopsy was entirely replaced with Hodgkin lymphoma associated with dense sclerosis (D). An EBER study shows scattered positive cells throughout the marrow (E), corresponding to the Hodgkin and Reed-Sternberg cells (F), which were CD30+ as illustrated (G). Hodgkin lymphoma infrequently involves the bone marrow in HIV-negative cases but some HIV+ patients can first present with extensive bone marrow disease (see box on HIV Hodgkin Lymphoma). (A courtesy Dr. Peter Pytel, University of Chicago.) A B CC E G D F
  • 8. Part VII  Hematologic Malignancies1252 comparable outcomes regardless of HIV status.71 In this study all HIV-seropositive patients were required to be on HAART to enroll, and the majority had CD4+ counts of greater than 200 cells/mm3 . Granulocyte colony-stimulating factor support was used throughout chemotherapy cycles, as well as PJP and other antimicrobial prophy- laxis. Differences in induction-related mortality, duration of neutro- penia, progression-free survival, or overall survival were not detected, although HIV-seropositive patients did have significantly more severe mucositis and infectious complications. Dose Most would agree that dose reduction is appropriate in patients with low CD4+ count (<100 cells/mm3 ), history of ongoing opportunistic infection, performance status below 75%, or compromised organ function. Some regimens begin with a 50% dose reduction in cyclo- phosphamide dose for CD4+ count of less than 100 cells/mm3 with a built-in dose escalation for the next cycle if well tolerated.64,68 Chemotherapy regimens of varying intensity depending on CD4+ count, performance status, and International Prognostic Index score have been studied, including a comparison of low-dose CHOP to standard-dose CHOP, with no differences found in overall survival based on the intensity of the chemotherapy regimen.66 In patients with very low CD4+ counts, low-dose chemotherapy is a reasonable treatment option, while maintaining the possibility of long-term disease-free survival in some. Antiretroviral Therapy A few key special issues in considering concurrent antiretroviral and lymphoma therapy include concerns about shared toxicities, drug-drug interactions, and risk for inability to comply with consis- tent antiretroviral dosing. Zidovudine is myelosuppressive and can exacerbate pancytopenia associated with lymphoma therapy. If patients are already on a zidovudine-containing regimen, it is generally possible to substitute an alternative regimen before the initiation of lymphoma therapy. Many antiretroviral agents alter the metabolism of drugs used in lymphoma treatment. This has been a particular concern when infusional chemotherapy regimens are used, and some investigators have chosen to stop chemotherapy before initiation of such regimens.64 However, in trials involving infusional chemotherapy that allowed patients already on a stable antiretroviral regimen to remain on that regimen during lymphoma treatment, major problems were not noted.68 Nausea and vomiting associated with chemotherapy regimens may interfere with regular antiviral dosing. Intermittent antiretroviral therapy raises concerns about the development of a resistant strain of HIV. Because of the concern that initiation of anti- retroviral therapy with cytotoxic chemotherapy might result in such resistance, many recommend delaying initiation of antiretroviral therapy until an appropriate regimen to control nausea and vomiting is established. Stopping antiretroviral therapy also carries with it some risks. When antiretroviral therapy includes drugs with different half- lives, stopping treatment may result in the longest-lived agent being present in the absence of other antiretroviral agents. This is particu- larly an issue for long-lived nonnucleoside reverse transcriptase inhibi- tors. Even a single dose of such agents in the absence of other antiretroviral agents may lead to resistance to that class of agents.Thus when an interruption of antiretroviral therapy is planned, specific strategies have been advocated, including a “staggered stop” or a change to a regimen with components that have similar half-lives.62 For patients already on antiretroviral treatment at the time of lymphoma diagnosis, the particulars of the regimen should be con- sidered during the pretreatment evaluation. Atazanavir and indinavir are associated with hyperbilirubinemia as a result of UGT1A1 inhibi- tion.62 This is an unconjugated hyperbilirubinemia similar to that occurring with Gilbert syndrome. Elevated total bilirubin in such patients is not indicative of hepatic involvement or other serious hepatic dysfunction and should not guide decisions about other settings. Positron emission tomography–computerized tomog- raphy (PET-CT), although useful, must also be interpreted with caution insofar as HIV infection itself, opportunistic infection, and immune reconstitution following the initiation of antiretroviral therapy are all associated with signal on metabolic imaging with fluorodeoxyglucose. CD4+ counts can help to guide evaluation insofar as CD4+ counts of greater than 300 cells/mm3 are typically associated with BL or HL,60 whereas these diagnoses would be unlikely in patients with very low CD4+ counts of less than 50 cells/mm3 (see box on AIDS Primary Central Nervous System Lymphoma).61 Treatment Aggressive chemotherapy for HIV-associated lymphoma was initially associated with morbidity and mortality related to immunocompro- mise. A phase III randomized study identified a reduced-dose regimen as preferable to standard dose.63 Lower doses were not associated with higher lymphoma cure rates but were associated with less chemotherapy-related morbidity and mortality. However, with the evolution of supportive care, including PJP prophylaxis and neutro- phil growth factors, tolerance of chemotherapy improved. With effec- tive antiretroviral therapy, long-term outcomes improved as well.64,65 And with full-dose therapies, some evidence emerged to suggest that stage-for-stage outcomes might be as good or better in patients with HIV-associated lymphoma compared to those without HIV.66 At the outset of therapy a series of questions must be addressed. Regimen Rituximab, cyclophosphamide, hydroxydaunomycin, vincristine, and prednisone (R-CHOP); and rituximab, infusional etoposide, predni- sone, vincristine, cyclophosphamide, and hydroxydaunomycin (R-EPOCH) have emerged as standard regimens.67,68 As in the treat- ment of DLBCL in patients without HIV infection, the value of infusional chemotherapy remains controversial. A recent pooled ret- rospective analysis favored the infusional regimen.69 However, insofar as the trials were conducted sequentially (R-CHOP between 1998 and 2002, and R-EPOCH between 2002 and 2006), it is difficult to exclude other factors such as the availability of better antiretroviral agents, improvements in supportive care, or changes in the popula- tion studied. However, there is general agreement that rituximab improves outcome, at least in patients with CD4+ counts of greater than 50 cells/mm3 . BL typically requires more intensive treatment regimens than those used for DLBCL.70 Thus there has been concern about using these regimens in the HIV-seropositive population. A small retrospec- tive study of cyclophosphamide, vincristine (Oncovin), doxorubicin, and methotrexate (CODOX-M)/ifosfamide, etoposide, and cytara- bine (IVAC) for BL that included 14 patients with HIV showed HIV-seropositive patients to have similar progression-free survival, overall survival, and complete response rates as compared to the HIV- negative BL patients. In a prospective Spanish study, HIV-seropositive and HIV-seronegative BL patients were treated with six cycles of intensive chemotherapy and rituximab and were found to have AIDS Primary Central Nervous System Lymphoma In acquired immunodeficiency syndrome (AIDS) primary central nervous system lymphoma (PCNSL), Epstein-Barr virus (EBV) polymerase chain reaction of the cerebrospinal fluid is positive approximately 90% of the time and rarely positive in patients with AIDS but without PCNSL. When EBV is detected in the cerebrospinal fluid of an AIDS patient, coupled with characteris- tic magnetic resonance imaging findings, this is sufficient to diagnose AIDS PCNSL without a confirmatory brain biopsy.
  • 9. Chapter 82  Virus-Associated Lymphoma 1253 whether infection of B cells plays any role in this process. A lym- phoma cell line that produces infectious HCV has been reported. Even in the absence of infection of B cells, interaction of the HCV E2 protein with CD81 on B cells may drive B-cell proliferation or lower the threshold for other B-cell stimuli to drive proliferation. Ig signaling may be activated by Ig-virus complexes, and Toll-like recep- tor 7 signaling may be activated by viral RNA. Finally, it is noted that E2 binding triggers expression of activation-induced deaminase, an enzyme that is important in generating somatic hypermutation and that has also been implicated in mediating mutations thought to play a role in DLBCL lymphomagenesis.77 Epidemiology of Viral Infection and Associated Lymphoma The association between HCV and lymphoma was first recognized in patients with HCV-associated type II mixed cryoglobulinemia, an autoimmune extrahepatic manifestation of HCV infection.79 There followed demonstration of an increased risk for certain subtypes of B-cell lymphoma (marginal zone lymphoma [MZL], lymphoplasma- cytic lymphoma and to a lesser extent DLBCL in HCV-infected patients).80-83 For example, in Taiwan the rate of chronic HCV infec- tion in patients with NHL was 11%, 10-fold higher than in the general Taiwanese population.84 Among HCV-infected patients with lymphoma, nodal and splenic MZL, but not mucosa-associated lym- phoid tissue (MALT) lymphomas, were increased. The HCV- lymphoma association is more apparent in some countries than others, with the association being established most clearly in Italy and Japan. A multitude of studies in regions or countries where HCV infection is less prevalent have failed to identify any association with lymphoma.85-87 Further evidence in support of an etiologic relationship comes from studies in which successful treatment of HCV was followed by lymphoma regression.88 The most dramatic illustration comes from patients with splenic lymphoma with villous lymphocytes treated with ribavirin and interferon. Certain HCV genotypes may confer increased risk for NHL, with genotypes 2a/III and 2b/IV seen more frequently in the HCV- seropositive patients that develop NHL. Diagnostic Considerations In contrast to EBV-, KSHV-, or HTLV-1–associated tumors, there is no established role for studies demonstrating HCV nucleic acid or protein in tumor cells. Thus serologic study and measurement of HCV copy number are the only tools available for inferring an asso- ciation. We recommend checking HCV serologic characteristics in all patients with B-cell lymphomas most commonly associated with chronic HCV infection.89 In addition, screening patients with chronic HCV for a monoclonal gammopathy and cryoglobulinemia may be of benefit to identify patients at highest risk for malignant transfor- mation. Elevated serum γ-globulin levels have been found to be a predictor of NHL among patients with type II mixed cryoglobuline- mia.90 In patients who are HCV seropositive, we evaluate HCV RNA in plasma. Therapy In patients with indolent lymphomas and untreated HCV infection, antiviral treatment may obviate the need for cytotoxic chemotherapy and should be considered as an initial therapeutic strategy. The studies showing that antiviral therapy may lead to regression of HCV- associated lymphomas involved treatment with ribavirin and inter- feron. In the last year new antiviral agents have become available, notably protease inhibitors specific for the HCV protease. Likelihood of response to older therapy is a function of viral genotype, host genetics (IL28R polymorphisms play a critical role in response to chemotherapy dose adjustments. Ritonavir inhibits the clearance of midazolam, phenytoin, and voriconazole and other agents metabo- lized by the cytochrome P-450 CYP3A4 pathway. Supportive Care Although PJP prophylaxis is recommended only when the CD4+ count is less than 200 cells/mm3 for HIV patients not receiving cytotoxic chemotherapy, prophylaxis is universally recommended for HIV patients receiving cytotoxic chemotherapy. The following are also commonly used: fungal prophylaxis with fluconazole; herpes simplex and varicella prophylaxis with acyclovir, valacyclovir, or fam- ciclovir; quinolone prophylaxis when neutrophil counts fall below 1000 cells/mm3 ; and granulocyte growth factors. Bone Marrow Transplantation in HIV Patients Autologous bone marrow transplant has been successful in HIV- seropositive NHL patients, with these patients having adequate stem cell mobilization, nonrelapse mortality rates comparable to those for HIV-negative patients, count recovery within 2 weeks of stem cell rescue, and maintained control of HIV viral loads and CD4+ counts after high-dose chemotherapy.73,74 There have also been successful reduced-intensity allogeneic bone marrow transplants in HIV- seropositive patients, making the possibilities for treating HIV patients with NHL even more vast, even in those patients with chemotherapy-resistant disease (see box on Autologous Bone Marrow Transplant in an HIV-Seropositive Hodgkin Lymphoma Patient).75 HEPATITIS C VIRUS Viral Biology and B-Lymphocyte Proliferation HCV is an enveloped positive-strand RNA virus.76,77 Infection involves interactions between E2, a viral structural protein with two hypervariable regions, and a cellular protein CD81 present on hepa- tocytes and B lymphocytes. A polyprotein is translated from viral RNA and is cleaved by cellular and viral proteases, including NS3, to yield proteins required for viral replication. The RNA-dependent RNA polymerase that replicates the viral genome lacks proofreading capacity, thus generating genetic heterogeneity among viral progeny. Viral replication occurs predominantly in the liver, but some evidence suggests that B cells may also be infected. Chronic infection can be associated with mixed cryoglobulinemia, a systemic vasculitis that results from clonal expansion of B cells producing an IgM autoantibody against IgG, leading to deposition of immune complexes on endothelial surfaces, resulting in inflamma- tion.78 Several hypotheses have been advanced with regard to how HCV might drive B-cell proliferation. There is controversy as to Autologous Bone Marrow Transplant in an HIV-Seropositive Hodgkin Lymphoma Patient A 38-year-old human immunodeficiency virus (HIV)–positive patient with CD4+ count of 485 cells/mm3 is diagnosed with classic Hodgkin lymphoma (HL) and treated with doxorubicin (Adriamycin), bleomycin, vinblastine, and dacarbazine (ABVD), achieving a complete remission. Two years later he presents with retroperitoneal lymphadenopathy and is found on biopsy to have relapsed HL. He is treated with salvage chemotherapy with com- plete response, as well as good performance status and no active infections. His HIV remains well controlled on antiretroviral therapy. He is deemed an excellent candidate for high-dose therapy and undergoes consolidation with autologous bone marrow transplant.
  • 10. Part VII  Hematologic Malignancies1254 Choi I, Tanosaki R, Uike N, et al: Long-term outcomes after hematopoietic SCT for adult T-cell leukemia/lymphoma: Results of prospective trials. Bone Marrow Transplant 46:116, 2011. Chuang SS, Liao YL, Chang ST, et al: Hepatitis C virus infection is signifi- cantly associated with malignant lymphoma in Taiwan, particularly with nodal and splenic marginal zone lymphomas. J Clin Pathol 63:595, 2010. Engels EA, Pfeiffer RM, Landgren O, et al: Immunologic and virologic predictors of AIDS-related non-Hodgkin lymphoma in the highly active antiretroviral therapy era. J Acquir Immune Defic Syndr 54:78, 2010. Epeldegui M, Hung YP, McQuay A, et al: Infection of human B cells with Epstein-Barr virus results in the expression of somatic hypermutation- inducing molecules and in the accrual of oncogene mutations. Mol Immunol 44:934, 2007. Evens AM, Roy R, Sterrenberg D, et al: Post-transplantation lymphoprolifera- tive disorders: Diagnosis, prognosis, and current approaches to therapy. Curr Oncol Rep 12:383, 2010. Giordano TP, Henderson L, Landgren O, et al: Risk of non-Hodgkin lym- phoma and lymphoproliferative precursor diseases in US veterans with hepatitis C virus. JAMA 297:2010, 2007. Guech-Ongey M, Simard EP, Anderson WF, et al: AIDS-related Burkitt lymphoma in the United States: What do age and CD4 lymphocyte pat- terns tell us about etiology and/or biology? Blood 116:5600, 2010. Heslop HE, Slobod KS, Pule MA, et al: Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 115:925, 2010. Hishizawa M, Kanda J, Utsunomiya A, et al: Transplantation of allogeneic hematopoietic stem cells for adult T-cell leukemia: A nationwide retrospec- tive study. Blood 116:1369, 2010. Ito M, Kusunoki H, Mochida K, et al: HCV infection and B-cell lymphoma- genesis. Adv Hematol 2011:835314, 2011. Jaffe ES, Campo E, Swerdlow SH, et al: The 2008 WHO classification of lymphoid neoplasms and beyond: Evolving concepts and practical applica- tions. Blood 117:5019, 2011. Keegan TH, Glaser SL, Clarke CA, et al: Epstein-Barr virus as a marker of survival after Hodgkin’s lymphoma: A population-based study. J Clin Oncol 23:7604, 2005. Kelly GL, Rickinson AB: Burkitt lymphoma: Revisiting the pathogenesis of a virus-associated malignancy. Hematology Am Soc Hematol Educ Program 2007:277, 2007. Kitahata MM, Achenbach CJ, Saag MS: Age at cancer diagnosis among persons with AIDS. Ann Int Med 154:642; author reply 643, 2011. Levine AM: HIV-associated lymphoma. Blood 115:2986, 2010. Libra M, Polesel J, Russo AE, et al: Extrahepatic disorders of HCV infection: A distinct entity of B-cell neoplasia? Int J Oncol 36:1331, 2010. Matsuoka M, Jeang KT: Human T-cell leukaemia virus type 1 (HTLV-1) infectivity and cellular transformation. Nat Rev Cancer 7:270, 2007. Matsuoka M, Jeang KT: Human T-cell leukemia virus type 1 (HTLV-1) and leukemic transformation: Viral infectivity, Tax, HBZ and therapy. Onco- gene 30:1379, 2011. Moore PS, Chang Y: KSHV: Forgotten but not gone. Blood 117:6973, 2011. Ratner L, Harrington W, Feng X, et al: Human T cell leukemia virus reactiva- tion with progression of adult T-cell leukemia-lymphoma. PLoS One 4:e4420, 2009. Rudek MA, Flexner C, Ambinder RF: Use of antineoplastic agents in patients with cancer who have HIV/AIDS. Lancet Oncol 12:905, 2011. Savoldo B, Goss JA, Hammer MM, et al: Treatment of solid organ transplant recipients with autologous Epstein Barr virus-specific cytotoxic T lympho- cytes (CTLs). Blood 108:2942, 2006. Sparano JA, Lee JY, Kaplan LD, et al: Rituximab plus concurrent infusional EPOCH chemotherapy is highly effective in HIV-associated B-cell non- Hodgkin lymphoma. Blood 115:3008, 2010. Uldrick TS, Polizzotto MN, Aleman K, et al: High-dose zidovudine plus valganciclovir for Kaposi sarcoma herpesvirus-associated multicentric Cas- tleman disease: A pilot study of virus-activated cytotoxic therapy. Blood 117:6977, 2011. Vereide DT, Sugden B: Lymphomas differ in their dependence on Epstein- Barr virus. Blood 117:1977, 2011. For complete list of references log on to www.expertconsult.com. protease inhibitors) and other factors. The field is rapidly evolving, and colleagues with specific expertise in appropriate antiviral approaches should be consulted.76 Rituximab has posed an interesting dilemma for the treatment of patients with HCV and lymphoma. It has been reported that HCV plasma RNA increases following rituximab treatment, and there is certainly the possibility that elimination of B cells for 3 to 18 months following treatment may compromise humoral responses to the evo- lution of HCV quasispecies. However, in studies to date overall survival is not inferior.91 Similarly, combination chemotherapy is safe in patients with HCV infection.92 Rituximab is specifically recom- mended for the treatment of HCV-associated cryoglobulinemia (although as in the treatment of Waldenström macroglobulinemia, it must be appreciated that the initial response to rituximab may be an increase in the IgM paraprotein level, necessitating plasmapheresis). Aspects of Therapy With regards to the use of rituximab to treat B-cell lymphomas in patients with HCV, overall survival is not inferior, although there do appear to be increased rates of hepatotoxicity and rises in HCV viral load during therapy. Similarly, combination chemotherapy is safe in patients with HCV infection, although HCV RNA levels can rise during treatment. Interestingly, patients with HCV and splenic MZL have had regression of their tumors with treatment for HCV infection with interferon-α and ribavirin, an effect not seen in HCV-negative patients with splenic MZL treated with the same regimen. FUTURE DIRECTIONS In this chapter a variety of virus-associated lymphomas and lympho- proliferative diseases have been reviewed. For most of these lympho- mas, standard antiviral drugs do not have a role in treatment. There are, however, several exceptions, and these are worth highlighting. Antiviral therapy for HCV-associated splenic lymphoma with villous lymphocytes is accepted as a standard approach and likely has a role in the treatment of other HCV-associated indolent lymphomas. Simi- larly, ganciclovir or valganciclovir appears to have a role in the man- agement of MCD associated with KSHV in HIV patients. And of course there is an established role for antiretroviral therapy in the treatment of HIV patients with malignancy. There are virus-targeted therapies that are broadly accepted as standard, including adoptive immunotherapy with EBV-specific T cells for PTLD. The use of targeted T cells also has promise in other settings, including EBV- associated HL. Other virus-targeted therapies are being developed. Some involve vaccination; others involve induction of viral genes in tumor cells, rendering them more susceptible to pharmacologic treat- ment. Finally, it may ultimately be possible to prevent some kinds of lymphoma by preventing viral infection or altering the host response to viral infection. SUGGESTED READINGS Balsalobre P, Diez-Martin JL, Re A, et al: Autologous stem-cell transplanta- tion in patients with HIV-related lymphoma. J Clin Oncol 27:2192, 2009. Barta SK, Lee JY, Kaplan LD, et al: Pooled analysis of AIDS malignancy consortium trials evaluating rituximab plus CHOP or infusional EPOCH chemotherapy in HIV-associated non-Hodgkin lymphoma. Cancer doi: 10.1002/cncr.26723. [Epub ahead of print], 2011. Bazarbachi A, Suarez F, Fields P, et al: How I treat adult T-cell leukemia/ lymphoma. Blood 118:1736, 2011. Bower M, Newsom-Davis T, Naresh K, et al: Clinical features and outcome in HIV-associated multicentric Castleman’s disease. J Clin Oncol 29:2481, 2011. Carbone A, Cesarman E, Spina M, et al: HIV-associated lymphomas and gamma-herpesviruses. Blood 113:1213, 2009.
  • 11. Chapter 82  Virus-Associated Lymphoma 1254.e1 Key Words Adult T-cell leukemia/lymphoma Burkitt lymphoma Epstein-Barr virus Hepatitis C virus Hodgkin lymphoma Human T-lymphotropic virus-1 Human immunodeficiency virus type 1 Kaposi sarcoma–associated herpesvirus (human herpesvirus 8) Marginal zone lymphoma Natural killer/T-cell lymphoma Posttransplant lymphoma Splenic lymphoma
  • 12. Part VII  Hematologic Malignancies1254.e2 REFERENCES 1. Cohen JI: Epstein-Barr virus infection. N Engl J Med 343:481, 2000. 2. Thorley-Lawson DA, Gross A: Persistence of the Epstein-Barr virus and the origins of associated lymphomas. N Engl J Med 350:1328, 2004. 3. Savoldo B, Goss JA, Hammer MM, et al: Treatment of solid organ transplant recipients with autologous Epstein Barr virus-specific cyto- toxic T lymphocytes (CTLs). Blood 108:2942, 2006. 4. Roughan JE, Torgbor C, Thorley-Lawson DA: Germinal center B cells latently infected with Epstein-Barr virus proliferate extensively but do not increase in number. J Virol 84:1158, 2010. 5. Mainou BA, Raab-Traub N: LMP1 strain variants: Biological and molec- ular properties. J Virol 80:6458, 2006. 6. Vereide DT, Sugden B: Lymphomas differ in their dependence on Epstein-Barr virus. Blood 117:1977, 2011. 7. Epeldegui M, Hung YP, McQuay A, et al: Infection of human B cells withEpstein-Barrvirusresultsintheexpressionofsomatichypermutation- inducing molecules and in the accrual of oncogene mutations. Mol Immunol 44:934, 2007. 8. Rovedo M, Longnecker R: Epstein-Barr virus latent membrane protein 2A preferentially signals through the Src family kinase Lyn. J Virol 82:8520, 2008. 9. Williams H, Crawford DH: Epstein-Barr virus: The impact of scientific advances on clinical practice. Blood 107:862, 2006. 10. Higgins CD, Swerdlow AJ, Macsween KF, et al: A study of risk factors for acquisition of Epstein-Barr virus and its subtypes. J Infect Dis 195:474, 2007. 11. Tierney RJ, Edwards RH, Sitki-Green D, et al: Multiple Epstein-Barr virus strains in patients with infectious mononucleosis: Comparison of ex vivo samples with in vitro isolates by use of heteroduplex tracking assays. J Infect Dis 193:287, 2006. 12. Glaser SL, Gulley ML, Borowitz MJ, et al: Inter- and intra-observer reliability of Epstein-Barr virus detection in Hodgkin lymphoma using histochemical procedures. Leuk Lymphoma 45:489, 2004. 13. Hsieh PP, Tung CL, Chan AB, et al: EBV viral load in tumor tissue is an important prognostic indicator for nasal NK/T-cell lymphoma. Am J Clin Patho 128:579, 2007. 14. MacMahon EM, Glass JD, Hayward SD, et al: Epstein-Barr virus in AIDS-related primary central nervous system lymphoma. Lancet 338:969, 1991. 15. Jaffe ES, Campo E, Swerdlow SH, et al: The 2008 WHO classification of lymphoid neoplasms and beyond: Evolving concepts and practical applications. Blood 117:5019, 2011. 16. Carbone A, Cesarman E, Spina M, et al: HIV-associated lymphomas and gamma-herpesviruses. Blood 113:1213, 2009. 17. Evens AM, Roy R, Sterrenberg D, et al: Post-transplantation lympho­ proliferative disorders: Diagnosis, prognosis, and current approaches to therapy. Curr Oncol Rep 12:383, 2010. 18. Murray PG, Deacon E, Young LS, et al: Localization of Epstein-Barr virus in Castleman’s disease by in situ hybridization and immunohisto- chemistry. Hematol Pathol 9:17, 1995. 19. Heslop HE, Slobod KS, Pule MA, et al: Long-term outcome of EBV- specific T-cell infusions to prevent or treat EBV-related lymphoprolifera- tive disease in transplant recipients. Blood 115:925, 2010. 20. Swinnen LJ, Costanzo-Nordin MR, Fisher SG, et al: Increased incidence of lymphoproliferative disorder after immunosuppression with the monoclonal antibody OKT3 in cardiac-transplant recipients. N Engl J Med 323:1723, 1990. 21. Glaser SL, Lin RJ, Stewart SL, et al: Epstein-Barr virus-associated Hodg- kin’s disease: Epidemiologic characteristics in international data. Int J Cancer 70:375, 1997. 22. Keegan TH, Glaser SL, Clarke CA, et al: Epstein-Barr virus as a marker of survival after Hodgkin’s lymphoma: A population-based study. J Clin Oncol 23:7604, 2005. 23. Hjalgrim H, Askling J, Rostgaard K, et al: Characteristics of Hodgkin’s lymphoma after infectious mononucleosis. N Engl J Med 349:1324, 2003. 24. Ambinder RF, Browning PJ, Lorenzana I, et al: Epstein-Barr virus and childhood Hodgkin’s disease in Honduras and the United States. Blood 81:462, 1993. 25. Glaser SL, Gulley ML, Clarke CA, et al: Racial/ethnic variation in EBV- positive classical Hodgkin lymphoma in California populations. Int J Cancer 123:1499, 2008. 26. Glaser SL, Clarke CA, Gulley ML, et al: Population-based patterns of human immunodeficiency virus-related Hodgkin lymphoma in the Greater San Francisco Bay Area, 1988-1998. Cancer 98:300, 2003. 27. Siebert JD, Ambinder RF, Napoli VM, et al: Human immunodeficiency virus-associated Hodgkin’s disease contains latent, not replicative, Epstein-Barr virus. Hum Pathol 26:1191, 1995. 28. Bollard CM, Aguilar L, Straathof KC, et al: Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin’s disease. J Exp Med 200:1623, 2004. 29. Kelly GL, Rickinson AB: Burkitt lymphoma: Revisiting the pathogenesis of a virus-associated malignancy. Hematology Am Soc Hematol Educ Program 2007:277, 2007. 30. Moore PS, Chang Y: KSHV: Forgotten but not gone. Blood 117:6973, 2011. 31. Ambinder RF, Cesarman E: Clinical and pathological aspects of EBV and KSHV infection. In: Arvin A, Campadelli-Fiume G, Mocarski E, et al, editors: Human herpesviruses: Biology, therapy, and immunoprophy- laxis, Cambridge, 2007, Cambridge University Press. 32. Cannon JS, Nicholas J, Orenstein JM, et al: Heterogeneity of viral IL-6 expression in HHV-8-associated diseases. J Infect Dis 180:824, 1999. 33. Du MQ, Liu H, Diss TC, et al: Kaposi sarcoma-associated herpesvirus infects monotypic (IgM lambda) but polyclonal naive B cells in Castle- man disease and associated lymphoproliferative disorders. Blood 97:2130, 2001. 34. Bower M, Newsom-Davis T, Naresh K, et al: Clinical features and outcome in HIV-associated multicentric Castleman’s disease. J Clin Oncol 29:2481, 2011. 35. Casper C: The aetiology and management of Castleman disease at 50 years: Translating pathophysiology to patient care. Br J Haematol 129:3, 2005. 36. Uldrick TS, Polizzotto MN, Aleman K, et al: High-dose zidovudine plus valganciclovir for Kaposi sarcoma herpesvirus-associated multicentric Castleman disease: A pilot study of virus-activated cytotoxic therapy. Blood 117:6977, 2011. 37. Matsuoka M, Jeang KT: Human T-cell leukemia virus type 1 (HTLV-1) and leukemic transformation: Viral infectivity, Tax, HBZ and therapy. Oncogene 30:1379, 2011. 38. Bazarbachi A, Suarez F, Fields P, et al: How I treat adult T-cell leukemia/ lymphoma. Blood 118:1736, 2011. 39. Bellon M, Baydoun HH, Yao Y, et al: HTLV-I Tax-dependent and -independent events associated with immortalization of human primary T lymphocytes. Blood 115:2441, 2010. 40. Matsuoka M, Jeang KT: Human T-cell leukaemia virus type 1 (HTLV-1) infectivity and cellular transformation. Nat Rev Cancer 7:270, 2007. 41. Sonoda S, Li HC, Tajima K: Ethnoepidemiology of HTLV-1 related diseases: Ethnic determinants of HTLV-1 susceptibility and its world- wide dispersal. Cancer Sci 102:295, 2011. 42. Koga Y, Iwanaga M, Soda M, et al: Trends in HTLV-1 prevalence and incidence of adult T-cell leukemia/lymphoma in Nagasaki, Japan. J Med Virol 82:668, 2010. 43. Ratner L, Harrington W, Feng X, et al: Human T cell leukemia virus reactivation with progression of adult T-cell leukemia-lymphoma. PLoS One 4:e4420, 2009. 44. Jiang L, Yuan CM, Hubacheck J, et al: Variable CD52 expression in mature T cell and NK cell malignancies: Implications for alemtuzumab therapy. Br J Haematol 145:173, 2009. 45. Morris J, Sharma K, O’Mahony D, et al: A phase II study of the efficacy and toxicity of alemtuzumab (Campath) for treatment of human T cell lymphotrophic virus-1 (HTLV-1)-associated adult T cell leukemia/ lymphoma (ATL). AIDS Res Hum Retroviruses 23:632, 2007. 46. Choi I, Tanosaki R, Uike N, et al: Long-term outcomes after hemato- poietic SCT for adult T-cell leukemia/lymphoma: Results of prospective trials. Bone Marrow Transplant 46:116, 2011. 47. Hishizawa M, Kanda J, Utsunomiya A, et al: Transplantation of alloge- neic hematopoietic stem cells for adult T-cell leukemia: A nationwide retrospective study. Blood 116:1369, 2010.
  • 13. Chapter 82  Virus-Associated Lymphoma 1254.e3 48. Epeldegui M, Vendrame E, Martinez-Maza O: HIV-associated immune dysfunction and viral infection: Role in the pathogenesis of AIDS-related lymphoma. Immunol Res 48:72, 2010. 49. Kitahata MM, Achenbach CJ, Saag MS: Age at cancer diagnosis among persons with AIDS. Ann Intern Med 154:642; author reply 643, 2011. 50. Shen L, Siliciano RF: Viral reservoirs, residual viremia, and the potential of highly active antiretroviral therapy to eradicate HIV infection. J Allergy Clin Immunol 122:22, 2008. 51. Gaidano G, Pasqualucci L, Capello D, et al: Aberrant somatic hypermu- tation in multiple subtypes of AIDS-associated non-Hodgkin lymphoma. Blood 102:1833, 2003. 52. Epeldegui M, Thapa DR, De la Cruz J, et al: CD40 ligand (CD154) incorporated into HIV virions induces activation-induced cytidine deaminase (AID) expression in human B lymphocytes. PloS one 5:e11448, 2010. 53. Dean M, Jacobson LP, McFarlane G, et al: Reduced risk of AIDS lym- phoma in individuals heterozygous for the CCR5-Δ32 mutation. Cancer Res 59:3561, 1999. 54. Goedert JJ: The epidemiology of acquired immunodeficiency syndrome malignancies. Semin Oncol 27:390, 2000. 55. Levine AM: HIV-associated lymphoma. Blood 115:2986, 2010. 56. Bohlius J, Schmidlin K, Costagliola D, et al: Incidence and risk factors of HIV-related non-Hodgkin’s lymphoma in the era of combination antiretroviral therapy: A European multicohort study. Antivir Ther 14:1065, 2009. 57. Bonnet F, Lewden C, May T, et al: Malignancy-related causes of death in human immunodeficiency virus-infected patients in the era of highly active antiretroviral therapy. Cancer 101:317, 2004. 58. Engels EA, Pfeiffer RM, Landgren O, et al: Immunologic and virologic predictors of AIDS-related non-Hodgkin lymphoma in the highly active antiretroviral therapy era. J Acquir Immune Defic Syndr 54:78, 2010. 59. Biggar RJ, Jaffe ES, Goedert JJ, et al: Hodgkin lymphoma and immu- nodeficiency in persons with HIV/AIDS. Blood 108:3786, 2006. 60. Guech-Ongey M, Simard EP, Anderson WF, et al: AIDS-related Burkitt lymphoma in the United States: What do age and CD4 lymphocyte patterns tell us about etiology and/or biology? Blood 116:5600, 2010. 61. Ambinder RF, Bhatia K, Martinez-Maza O, et al: Cancer biomarkers in HIV patients. Curr Opin HIV AIDS 5:531, 2010. 62. Rudek MA, Flexner C, Ambinder RF: Use of antineoplastic agents in patients with cancer who have HIV/AIDS. Lancet Oncol 12:905, 2011. 63. Kaplan LD, Straus DJ, Testa MA, et al: Low-dose compared with standard-dose m-BACOD chemotherapy for non-Hodgkin’s lymphoma associated with human immunodeficiency virus infection. National Insti- tute of Allergy and Infectious Diseases AIDS Clinical Trials Group. N Engl J Med 336:1641, 1997. 64. Little RF, Pittaluga S, Grant N, et al: Highly effective treatment of acquired immunodeficiency syndrome-related lymphoma with dose- adjusted EPOCH: Impact of antiretroviral therapy suspension and tumor biology. Blood 101:4653, 2003. 65. Ratner L, Lee J, Tang S, et al: Chemotherapy for human immunodefi- ciency virus-associated non-Hodgkin’s lymphoma in combination with highly active antiretroviral therapy. J Clin Oncol 19:2171, 2001. 66. Mounier N, Spina M, Gabarre J, et al: AIDS-related non-Hodgkin lymphoma: Final analysis of 485 patients treated with risk-adapted inten- sive chemotherapy. Blood 107:3832, 2006. 67. Kaplan LD, Lee JY, Ambinder RF, et al: Rituximab does not improve clinical outcome in a randomized phase 3 trial of CHOP with or without rituximab in patients with HIV-associated non-Hodgkin lymphoma: AIDS-Malignancies Consortium Trial 010. Blood 106:1538, 2005. 68. Sparano JA, Lee JY, Kaplan LD, et al: Rituximab plus concurrent infu- sional EPOCH chemotherapy is highly effective in HIV-associated B-cell non-Hodgkin lymphoma. Blood 115:3008, 2010. 69. Barta SK, Lee JY, Kaplan LD, et al: Pooled analysis of AIDS malignancy consortium trials evaluating rituximab plus CHOP or infusional EPOCH chemotherapy in HIV-associated non-Hodgkin lymphoma. Cancer doi: 10.1002/cncr.26723. [Epub ahead of print], 2011. 70. Levine AM: Management of AIDS-related lymphoma. Curr Opin Oncol 20:522, 2008. 71. Oriol A, Ribera JM, Bergua J, et al: High-dose chemotherapy and immu- notherapy in adult Burkitt lymphoma: Comparison of results in human immunodeficiency virus-infected and noninfected patients. Cancer 113:117, 2008. 72. Gopal S, Martin K, Richards K, et al: Clinical presentation, treatment, and outcomes among 65 patients with HIV-associated lymphoma treated at the University of North Carolina, 2000-2010. AIDS Res Hum Retro- viruses [Epub ahead of print] 2011. 73. Balsalobre P, Diez-Martin JL, Re A, et al: Autologous stem-cell trans- plantation in patients with HIV-related lymphoma. J Clin Oncol 27:2192, 2009. 74. Gabarre J, Marcelin AG, Azar N, et al: High-dose therapy plus autolo- gous hematopoietic stem cell transplantation for human immunodefi- ciency virus (HIV)-related lymphoma: Results and impact on HIV disease. Haematologica 89:1100, 2004. 75. Bryant A, Milliken S: Successful reduced-intensity conditioning alloge- neic HSCT for HIV-related primary effusion lymphoma. Biol Blood Marrow Transplant 14:601, 2008. 76. Rosen HR: Clinical practice. Chronic hepatitis C infection. N Engl J Med 364:2429, 2011. 77. Ito M, Kusunoki H, Mochida K, et al: HCV infection and B-cell lym- phomagenesis. Adv Hematol 2011:835314, 2011. 78. Della Rossa A, Baldini C, Tavoni A, et al: How HCV has changed the approach to mixed cryoglobulinemia. Clin Exp Rheumatol 27:S115, 2009. 79. Monti G, Pioltelli P, Saccardo F, et al: Incidence and characteristics of non-Hodgkin lymphomas in a multicenter case file of patients with hepatitis C virus-related symptomatic mixed cryoglobulinemias. Arch Intern Med 165:101, 2005. 80. Anderson LA, Engels EA: Hepatitis C virus infection and non-Hodgkin lymphoma: Interesting association or causal relationship? Int J Cancer 122:x, 2008. 81. Giordano TP, Henderson L, Landgren O, et al: Risk of non-Hodgkin lymphoma and lymphoproliferative precursor diseases in US veterans with hepatitis C virus. JAMA 297:2010, 2007. 82. Libra M, Polesel J, Russo AE, et al: Extrahepatic disorders of HCV infec- tion: A distinct entity of B-cell neoplasia? Int J Oncol 36:1331, 2010. 83. Amin J, Dore GJ, O’Connell DL, et al: Cancer incidence in people with hepatitis B or C infection: A large community-based linkage study. J Hepatol 45:197, 2006. 84. Chuang SS, Liao YL, Chang ST, et al: Hepatitis C virus infection is significantly associated with malignant lymphoma in Taiwan, particularly with nodal and splenic marginal zone lymphomas. J Clin Pathol 63:595, 2010. 85. Varma S, Menon MC, Garg A, et al: Hepatitis C virus infection among patients with non-Hodgkin’s lymphoma in northern India. Hepatol Int 5:688, 2011. 86. Omland LH, Jepsen P, Krarup H, et al: Liver cancer and non-Hodgkin lymphoma in hepatitis C virus-infected patients: Results from the DANVIR cohort study. Int J Cancer 130:2310, 2012. 87. Isikdogan A, Ayyildiz O, Dursun M, et al: Hepatitis C virus in patients with non-Hodgkin’s lymphoma in southeastern Anatolia region of Turkey: A prospective case-control study of 119 patients. Leuk Lymphoma 44:1745, 2003. 88. Hermine O, Lefrere F, Bronowicki JP, et al: Regression of splenic lym- phoma with villous lymphocytes after treatment of hepatitis C virus infection. N Engl J Med 347:89, 2002. 89. Awasthi NP, Kashyap R, Kumari N, et al: Sjogren’s syndrome compli- cated by non-Hodgkin’s lymphoma: Spectrum of lymphoid prolifera- tions and associated hepatitis C virus infection. Indian J Pathol Microbiol 54:430, 2011. 90. Geri G, Terrier B, Semoun O, et al: Surrogate markers of B cell non- Hodgkin’s lymphoma in patients with hepatitis C virus-related cryo- globulinaemia vasculitis. Ann Rheum Dis 69:2177, 2010. 91. Ennishi D, Maeda Y, Niitsu N, et al: Hepatic toxicity and prognosis in hepatitis C virus-infected patients with diffuse large B-cell lymphoma treated with rituximab-containing chemotherapy regimens: A Japanese multicenter analysis. Blood 116:5119, 2010. 92. Ennishi D, Terui Y, Yokoyama M, et al: Monitoring serum hepatitis C virus (HCV) RNA in patients with HCV-infected CD20-positive B-cell lymphoma undergoing rituximab combination chemotherapy. Am J Hematol 83:59, 2008.