Small Lymphocytic Lymphoma (SLL).pdf- Ventoxen

ventoxenvenclexta 8 views 6 slides Oct 27, 2025
Slide 1
Slide 1 of 6
Slide 1
1
Slide 2
2
Slide 3
3
Slide 4
4
Slide 5
5
Slide 6
6

About This Presentation

Ventoxen is a generic version of VENCLEXTA which is a targeted cancer drug used to treat chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL), and acute myeloid leukemia (AML) in certain adults


Slide Content

Small Lymphocytic Lymphoma
Small Lymphocytic Lymphoma (SLL) is a type of non-Hodgkin lymphoma characterized by the
abnormal proliferation of mature B-lymphocytes, a subset of white blood cells crucial for immune
function. This disease is indolent, meaning it progresses slowly over time, often without
immediate aggressive symptoms. SLL is closely related to Chronic Lymphocytic Leukemia
(CLL), and in fact, the two are considered manifestations of the same underlying malignancy,
differing primarily in their anatomical presentation. While CLL predominantly involves the blood
and bone marrow, leading to elevated lymphocyte counts in the bloodstream, SLL is defined by
the accumulation of these abnormal cells mainly in the lymph nodes, spleen, and other lymphoid
tissues, with fewer than 5,000 monoclonal lymphocytes per cubic millimeter in the peripheral
blood. This distinction is more of a clinical classification than a biological difference, as the
cancer cells in both conditions appear identical under microscopic examination and share the
same genetic and immunophenotypic features.​


The World Health Organization (WHO) classifies SLL as a mature B-cell neoplasm within the
category of chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), emphasizing
their unity as a single entity. This unified approach has significant implications for diagnosis,
treatment, and prognosis, as strategies developed for CLL are directly applicable to SLL. The
disease typically affects older adults, with a median age at diagnosis around 65-70 years, and
shows a slight male predominance. In the United States, SLL accounts for approximately 6-7%
of all non-Hodgkin lymphomas, with an incidence rate of about 4.6 cases per 100,000 people
annually, though rates have been declining slightly over the past decade.

Understanding SLL requires appreciating its indolent nature: many patients live with the disease
for years or even decades without requiring treatment, entering a "watch and wait" phase where
regular monitoring suffices. However, in some cases, the disease can transform into a more
aggressive form, such as Richter's syndrome, which occurs in about 2-10% of patients and
involves evolution to diffuse large B-cell lymphoma or other high-grade lymphomas. This
transformation markedly worsens prognosis and necessitates prompt intervention. The impact of SLL extends beyond physical health, affecting quality of life through symptoms
like fatigue and recurrent infections, as well as psychological burdens from living with a chronic
cancer diagnosis. Advances in targeted therapies have revolutionized management, shifting
from traditional chemotherapy to more precise, less toxic options. This detailed discussion will
cover epidemiology, pathophysiology, clinical presentation, diagnostic approaches, staging,
treatment modalities, prognosis, ongoing research, and patient resources, providing a thorough,
accurate, and usable guide for patients, caregivers, and healthcare professionals.​






Epidemiology and Risk Factors
Epidemiologically, SLL is most prevalent in Western countries, with higher rates among
Caucasians compared to African Americans or Asians. The annual incidence in the U.S. is
estimated at 4-5 cases per 100,000, with a lifetime risk of about 1 in 175 for men and 1 in 230
for women. Age is the strongest risk factor; incidence rises sharply after age 50, peaking in the
70s. Familial predisposition plays a role, with first-degree relatives of affected individuals having
a 2-8-fold increased risk, suggesting genetic susceptibility. Genome-wide association studies
have identified polymorphisms in genes like BCL2, IRF4, and HLA regions that may contribute
to this heritability.
Environmental factors are less clearly defined but include exposure to certain chemicals, such
as herbicides and pesticides, which have been linked in agricultural workers. Chronic immune
stimulation, as seen in autoimmune diseases like rheumatoid arthritis or Sjögren's syndrome,
may also elevate risk, possibly through persistent B-cell activation. Viral infections, including
hepatitis C, have shown associations in some cohorts, though causality remains unproven.
Notably, unlike some lymphomas, SLL is not strongly associated with HIV or Epstein-Barr virus. Monoclonal B-cell lymphocytosis (MBL), a precursor condition where low levels of clonal B-cells
are detected in the blood without meeting lymphoma criteria, precedes SLL/CLL in many cases.
High-count MBL (greater than 0.5 x 10^9/L clonal cells) progresses to CLL/SLL at a rate of 1-2%
per year, serving as a screening opportunity in high-risk populations. Global variations exist;
lower rates in Asia may reflect genetic differences or underdiagnosis.

Recent data from 2024-2025 indicate stable incidence but improved survival due to better
diagnostics and treatments. Public health efforts focus on awareness in aging populations, as
early detection through routine blood work can identify asymptomatic cases.
Pathophysiology and Molecular Biology
At the cellular level, SLL arises from the clonal expansion of mature CD5-positive
B-lymphocytes that fail to undergo apoptosis, leading to their accumulation. These cells express
surface markers like CD19, CD20, CD23, and dim CD5, with low levels of surface
immunoglobulin, distinguishing them from other B-cell lymphomas. The pathophysiology
involves dysregulation of key signaling pathways, including the B-cell receptor (BCR) pathway,
which promotes survival and proliferation. Genetic aberrations are central: deletion of 13q14, containing microRNAs miR-15a and
miR-16-1 that regulate BCL2, is the most common (50-60%), leading to BCL2 overexpression
and apoptosis resistance. Trisomy 12 (15-20%) affects cell cycle regulation, while deletions in
11q (ATM gene) and 17p (TP53) (10-15%) confer poor prognosis by impairing DNA repair and
tumor suppression. Mutations in NOTCH1, SF3B1, and BIRC3 further disrupt signaling and
splicing. The bone marrow microenvironment plays a supportive role, with nurse-like cells and stromal
elements providing survival signals via CXCL12 and BAFF. In SLL, lymph node involvement
predominates, with pseudofollicular proliferation centers where cells proliferate under T-cell and
cytokine influence. Compared to CLL, SLL may have subtle differences, such as higher trisomy
12 incidence and lower 13q deletions, potentially explaining nodal tropism.
Immunosuppression arises from dysfunctional B-cells impairing T-cell function and
hypogammaglobulinemia, increasing infection risk. Autoimmune complications, like hemolytic
anemia or thrombocytopenia, occur in 10-25% of cases due to aberrant immune regulation.
Recent molecular insights from 2024-2025 include the role of epigenetic modifiers and
non-coding RNAs in disease progression, paving the way for novel therapies.
Clinical Presentation and Symptoms
SLL often presents insidiously, with many patients asymptomatic at diagnosis, discovered
incidentally via lymphadenopathy on physical exam or imaging. The hallmark symptom is
painless, persistent swelling of lymph nodes in the neck, axilla, or groin, which may fluctuate but
generally enlarges over months to years. Systemic "B symptoms" – fever, night sweats, and
unintentional weight loss (>10% body weight in 6 months) – occur in 10-20% and indicate more
advanced disease. Fatigue is ubiquitous, stemming from anemia or cytokine release, affecting daily activities.
Splenomegaly can cause abdominal fullness or early satiety, while hepatomegaly is less

common. Recurrent infections, particularly respiratory or skin, result from immune compromise.
Autoimmune cytopenias manifest as pallor (anemia), bruising (thrombocytopenia), or jaundice
(hemolysis).
Rare extranodal involvement includes skin (leukemia cutis), gastrointestinal tract, or central
nervous system, presenting with rashes, bleeding, or neurological deficits. In advanced stages,
bulky disease may compress structures, causing superior vena cava syndrome or ureteral
obstruction.
Differentiating SLL from CLL clinically: SLL lacks significant lymphocytosis (<5,000/μL), focusing
symptoms on nodal enlargement rather than blood-related issues. However, overlap is common,
and diagnoses can shift with disease evolution.
Diagnostic Approaches
Diagnosis of SLL requires a multifaceted approach, starting with history and physical
examination to identify lymphadenopathy or splenomegaly. Peripheral blood smear may show
small, mature lymphocytes with smudge cells, but lymphocytosis is minimal.
Flow cytometry on blood or tissue is pivotal, confirming the immunophenotype: CD5+, CD19+,
CD20 dim, CD23+, FMC7-, and cyclin D1- (distinguishing from mantle cell lymphoma). Lymph
node biopsy is essential for SLL, revealing diffuse effacement with proliferation centers
(pseudofollicles) of prolymphocytes and paraimmunoblasts. Immunohistochemistry supports the
diagnosis, with LEF1 positivity aiding differentiation.
Genetic testing via fluorescence in situ hybridization (FISH) detects chromosomal abnormalities
like del(17p) or del(11q), guiding prognosis and therapy. Next-generation sequencing identifies
TP53, ATM, or NOTCH1 mutations. Bone marrow biopsy assesses involvement, though not
always necessary for SLL.
Imaging with CT or PET-CT evaluates disease extent, though PET is less useful in indolent
lymphomas due to low FDG avidity. Differential diagnosis includes other small B-cell lymphomas
(follicular, marginal zone), reactive hyperplasia, or infections.
In 2025, liquid biopsy for circulating tumor DNA offers non-invasive monitoring.
Staging Systems
Staging for SLL follows the Ann Arbor system for lymphomas, modified by Lugano criteria.
●​Stage I: Involvement of a single lymph node region or extralymphatic site.
●​Stage II: Two or more lymph node regions on the same side of the diaphragm.
●​Stage III: Lymph node regions on both sides of the diaphragm.
●​Stage IV: Diffuse involvement of bone marrow, liver, or other extralymphatic organs.

"B" denotes systemic symptoms; "A" absence. Bulky disease (>10 cm) is noted.
For prognostic purposes, the CLL International Prognostic Index (CLL-IPI) is adapted,
incorporating age, stage, TP53 status, IGHV mutation, and beta-2 microglobulin levels,
stratifying into low, intermediate, high, and very high-risk groups. This predicts time to first
treatment and overall survival.
Recent updates in 2024-2025 emphasize integrating molecular markers for personalized
staging.
Treatment Options
Treatment for SLL is individualized, often starting with active surveillance ("watch and wait") for
asymptomatic, early-stage disease, as early intervention does not improve survival. Indications
for therapy include progressive lymphadenopathy, cytopenias, B symptoms, or rapid lymphocyte
doubling time.
First-line options have shifted to targeted agents. Bruton tyrosine kinase (BTK) inhibitors like
ibrutinib, acalabrutinib, or zanubrutinib disrupt BCR signaling, achieving high response rates
(80-90%) with continuous therapy. BCL2 inhibitors like venetoclax, often combined with
anti-CD20 monoclonal antibodies (rituximab or obinutuzumab), offer fixed-duration regimens,
inducing deep remissions. For example, venetoclax-obinutuzumab is approved for frontline use,
with progression-free survival exceeding 80% at 3 years. In relapsed/refractory settings, PI3K inhibitors (idelalisib, duvelisib) or next-generation BTKi
(pirtobrutinib for covalent BTKi-resistant cases) are options. CAR-T cell therapy, like
lisocabtagene maraleucel, shows promise for high-risk patients, with ongoing trials in 2025.
Chemotherapy (e.g., fludarabine-cyclophosphamide-rituximab) is reserved for young, fit patients
without TP53 aberrations, but its use has declined due to toxicity.
Supportive care includes vaccinations (pneumococcal, influenza), immunoglobulin replacement
for hypogammaglobulinemia, and prophylaxis against Pneumocystis jirovecii and herpesviruses
during therapy. For autoimmune complications, corticosteroids or rituximab are used.
2024-2025 consensus recommends sequencing based on prior therapy and mutations: BTKi
first for most, venetoclax for BTKi failure, and CAR-T for double-refractory disease. Radiation
therapy is limited to palliative use for bulky nodes.
Prognosis and Survival
Prognosis for SLL is generally favorable, with a 5-year relative survival rate of approximately
88% based on 2013-2019 data, improving to over 90% with modern therapies. Median overall
survival exceeds 10 years, and many patients die from unrelated causes. Favorable factors
include mutated IGHV, isolated 13q deletion, low CLL-IPI score, and early stage. Adverse

predictors: unmutated IGHV, del(17p)/TP53 mutation, complex karyotype, advanced age, and
comorbidities.
Transformation to Richter's syndrome reduces survival to months without aggressive treatment.
Recent advances have mitigated poor-risk features; e.g., venetoclax-based regimens improve
outcomes in TP53-mutated cases.
Quality of life is key, with long-term survivors facing risks of secondary malignancies (e.g., skin
cancer) and cardiovascular events from therapies. Regular follow-up every 3-6 months includes
exams, blood work, and imaging as needed.
Ongoing Research and Future Directions
Research in 2024-2025 focuses on combination therapies to achieve minimal residual disease
(MRD) negativity, potentially curative. Trials like CLL17 compare continuous BTKi vs.
fixed-duration venetoclax-ibrutinib. Bispecific antibodies (e.g., epcoritamab) and novel CAR-T
constructs target CD19/CD20 for refractory disease.
Epigenetic modulators and immune checkpoint inhibitors are explored, especially for Richter's
transformation. AI-driven prognostication and liquid biopsies for MRD monitoring are emerging.
Preventive strategies target MBL progression.
Global collaborations, like the Lymphoma Research Foundation workshops, guide
evidence-based updates.
Patient Resources and Support
Patients should access reputable organizations: CLL Society (cllsociety.org) for education and
support groups. Lymphoma Action (lymphoma-action.org.uk) offers CLL/SLL-specific info.
NCCN guidelines (nccn.org) provide patient versions. Clinical trials via clinicaltrials.gov,
especially UCSD for SLL.