The Immunosuppressed Patient


CD4 count (cell/ml)

Opportunistic infection

Primary regimen

<250

Coccidioidomycosis

Fluconazole/itraconazole

<200

Pneumocystis pneumonia

TMP-SMX

Histoplasmosis

Amphotericin B

<150

H. capsulatum

Itraconazole

<100

T. gondii encephalitis

Pyrimethamine

Cryptococcal meningitis

Amphotericin B

CMV retinitis

Ophthalmology consult

<50

M. tuberculosis

INH + RIF + PZA + EMB

MAC

Clarithromycin/azithromycin

Candida spp

Fluconazole

Aspergillosis

Voriconazole

CMV esophagitis/colitis

Ganciclovir

HPV warts

Podophyllotoxin, imiquimod, sinecatechin


CMV Cytomegalovirus; MAC Mycobacterium avium complex; INH Isoniazid; RIF Rifampin; PZA Pyrazinamide; EMB Ethambutol



Another consideration in the management of the HIV-positive patient is the development of multiorgan dysfunction associated with the infection. For instance, chronic vascular inflammation and elevated cytokine levels leads to cardiovascular disease and thrombosis [4]. HIV nephropathy and drug-induced kidney damage is a known cause of chronic kidney disease, and transplantation has emerged as a viable option [5]. Furthermore, direct invasion of gastrointestinal (GI) tract affects GI motility. Coviral infection with HPV increases incidence of anogenital disease. Clostridium difficile colitis is of special concern in this population due to chronic antimicrobial use affecting the GI flora. Chronic liver disease due to associated HBV/HCV infection, steatosis, and hepatocellular carcinoma has emerged as the leading cause of morbidity and mortality [6]. Liver transplantation is considered a possible alternative for liver failure in this population. Additionally, antiviral drugs affect proper endocrine function yielding insulin resistance, dyslipidemia, and lipodystrophy [6]. This multitude of disorders experienced by HIV infection often confounds perioperative management.

The side effect profile from the antiviral regimen is profound. Drug therapy often utilizes a combination of medications addressing different stages of the HIV infection pathway [7]. Initiation of ART leads to replenishment of CD4+ count with associated viral suppression [8]. Some patients develop an immune reconstitution inflammatory syndrome (IRIS) characterized by acute local and systemic inflammatory that is associated with clinical exacerbation [9]. This reaction has been linked to antigenic load of preexisting illnesses and opportunistic infections [9]. Dose- and agent-related toxicities of ART are numerous implicating mitochondrial dysfunction, metabolic derangements, myelosuppression, and allergic reactions [10]. For a summary of adverse effects with surgical implications refer to Table 22.2. Of particular importance to the surgeon is the direct nephrotoxic, hepatotoxic, and myelosuppression effects that may affect preoperative planning.


Table 22.2
Side effect of antiretroviral therapy

























Mechanism

Side effect

Mitochondrial dysfunction

Lactic acidosis, hepatic toxicity, pancreatitis, peripheral neuropathy, cardiomyopathy, pancreatitis, hepatic steatosis, lipodystrophy, proximal myopathy, polymyositis

Metabolic abnormalities

Fat maldistribution (lipodystrophy), dyslipidemia, hyperglycemia, glucose intolerance, insulin resistance, osteopenia, osteoporosis, osteonecrosis, arthritis

Bone marrow suppression

Anemia, neutropenia, thrombocytopenia

Allergic reactions

Rashes, hypersensitivity responses

Immune reconstitution inflammatory syndrome

Mycobacterium avium abscesses, Mycobacterium leprae lesions, sarcoidosis, Grave’s disease, PML, SLE, antiphospholipid syndrome, vasculitis, primary biliary cirrhosis, hepatitis, polymyositis, uveitis, ITP


ITP Immune thrombocytic purpura; PML Progressive multifocal leukoencephalopathy; SLE Systemic lupus erythematosus

Surgical indications in HIV/AIDS patients include elective and urgent indications as would be for any particular disease, as well as for HIV disease complications intended for prophylaxis or palliation. Malignancies more prevalent in HIV population includes Kaposi’s sarcoma or non-Hodgkin’s lymphoma and opportunistic infection such as cytomegalovirus (CMV), cryptosporidium, and mycobacterium avium complex (MAC) are well-documented causes of bowel obstruction, perforation, and peritonitis [11]. Additionally, anorectal pathologies such as hemorrhoids, ulcers, fissures, and anal condyloma occur relatively more frequently and palliative surgery can be performed safely. However, surgery should be individualized due to decreased wound healing rates. Prevention of anorectal disease is encouraged with HPV vaccination, conservative management, and avoidance of anorectal trauma or injury. When concerns for intraepithelial neoplasia are present, a biopsy helps identify HPV serotype to guide management [12].

Active HIV virus replication has distinct effects on surgical outcomes. Patients with CD4 count <200 cells/μl or viral load exceeding 30,000 copies/ml experience higher rate of complications [13]. Preoperative hypoalbuminemia (<3.5 g/dl), age, and decreased functional status have been associated with an increased 30-day postsurgical mortality among HIV patients [14]. These patients tend to be nutritionally deficient, chronically ill, and in poor state of health. Surprisingly, elective procedures for low-risk surgeries such as hernia repair, appendectomy, and cholecystectomy do not require extensive perioperative planning and can be performed safely [13]. Emergent surgery has higher mortality and complication rates due to impaired wound healing and surgical site infections [15].

Preoperative assessment of HIV patients requires an assessment of HIV disease status, medication history, nutritional, and functional status. Preoperative laboratories should include complete blood count (CBC) and comprehensive metabolic panel (CMP) to rule out myelosuppression, HIV nephropathy, or coagulopathy. Maximizing ART to improve viral suppression and CD4 counts is of particular importance and has to be addressed in preoperative planning, but caution must be employed if IRIS occurs.

In summary, HIV is a complex multisystem disease with multiple considerations affecting surgical evaluation and outcomes. Antiretroviral treatment has dramatically delayed disease progression; however, the side effect profile of these drugs, as well as complications of patients with chronic immunosuppression secondary to HIV, presents various challenges in the surgical setting. Low-risk and elective surgeries appear to present no additional risks, while emergent and high-risk surgeries endure complications mainly dependent on the individual’s state of health. Improvement of CD4 count and reduction of viral load with ART are effective methods to reduce complication rates.



Chemotherapy


Neoplastic disease is one of the leading causes of death worldwide with an incidence of 14 million in 2012, resulting in 8.2 million deaths [16]. Oncological management is complex and multimodal, often requiring combination of chemotherapy, radiation, and surgical intervention for definitive or palliative treatment. Aside from their role in solid tumor diagnosis and management, surgeons manage acute surgical interventions either separate from or influenced by the primary disease. Thus, basic knowledge of chemotherapy and its effect on immunosuppression is of utmost importance.

Chemotherapy ideally reduces disease burden by decreasing tumor size, halting disease progression, and abolishing micrometastases. In general, antineoplastic drugs act at the cellular level to reduce rapidly proliferating cells [17]. As a result, these drugs damage healthy cells with similar characteristics residing in the bone marrow, gastrointestinal tract (GI) and skin tissue. Immunosuppression and myelosuppression are of particular interest in perioperative management. This section will focus on common perioperative presentations and surgical implications of side effects encountered during antineoplastic therapy.

Chemotherapy agents are divided into alkylating agents, antitumor antibiotics, antimetabolites, plant alkaloids, hormonal therapy, and targeted tissue therapy [17]. Many of these agents function to inhibit the cell cycle in a phase-specific (antimetabolite, vinca alkaloid, and bleomycin) or non-specific manner (alkylating agents and antibiotics) [17]. A summary of side effects is found in Table 22.3.


Table 22.3
Chemotherapy agents and their common adverse effects































































































































































Class

Agent

Adverse effects

Alkylating agent

Nitrosurea

Carmustine, lomustine

Pulmonary fibrosis

Methylating agent

Procarbazine

Edema, tachycardia

Dacarbazine

Hepatic necrosis, obstruction and hepatic vein obstruction

Temozolomide

Seizure, gait abnormality, peripheral edema

Platinum

Cisplatin

Acute renal tubular necrosis

Carboplatin

Magnesium wasting

Oxaliplatin

Peripheral sensory neuropathy, ototoxicity

Nitrogen mustard

Cyclophophomide

Pericarditis

Ifosfomide

Pericardial effusions, pulmonary fibrosis, hemorrhagic cystitis, anemia, edema

Melphalan

SIADH

Chlorambucil

SIADH, seizure

Antimetabolite

Anthracycline/anthraquinolone

Doxorubicin

Cardiomyopathy

Daunorubicin, epirubicin, idarubicin, mitoxantrone

Electrocardiogram changes

Antitumor antibiotic

Bleomycin

Pulmonary fibrosis

Mitomycin C

Pneumonitis, pulmonary hypertension

Antimetabolite: pyrimidine analogue

Capecitabine

Myocardial ischemia

Cytarabine

Coronary vasospasm

5-Fluorouracil
 

Gemcitabine

Proteinuria

Antimetabolite: purine analogue

Pentostatin

Pulmonary toxicity, deep vein thrombosis, chest pain, arrhythmia, AV block

Thioguanine

Hepatotoxicity

Cladribine

Thrombosis, tachycardia, ARF, tumor lysis syndrome

Fludarabine

CVA, TIA, CHF, arrhythmia, ARF, tumor lysis syndrome

Mercaptopurine

Intrahepatic cholestasis and centrilobular necrosis

Antimetabolite: folate antagonist

Methotrexate

Elevated LFTs, pulmonary edema, pleural effusions, meningismus, encephalopathy, myelosuppression

Urea substitute

Hydroxyurea

Seizure, edema

Microtubule assembly inhibitor

Taxane

Paclitaxel

Peripheral neuropathy

Docetaxel

Bradycardia, autonomic dysfunction

Alkaloid

Vinblastine

Hypertension, MI, CVA, Raynaud’s phenomenon, SIADH, GI bleeding

Vincristine

Paresthesia, laryngeal nerve palsy, autonomic dysfunction, orthostasis, SIADH

Vascular endothelial growth factor inhibitor

Tyrosine kinase

Imatinib

Edema, LV dysfunction

Sorafenib

Hypertension, MI

Sunitinib

MI, thromboembolism, adrenal insufficiency, hypothyroidism, cardiomyopathy, QT prolongation and torsade de pointes

Dasatinib

Edema, cardiomyopathy, QT prolongation, pulmonary hemorrhage, platelet dysfunction

Nilotinib

QT prolongation, edema, hypertension

Epidermal growth factors receptor inhibitor
 
Erlotinib

DVT, CVA, MI, arrhythmia, pulmonary toxicity, syncope, edema

Lapatinib

Cardiomyopathy, QT prolongation, pulmonary hemorrhage

Panitunab

Pulmonary fibrosis and edema

Angiogenesis inhibitor

Immunomodulation

Thalidomide

Thromboembolism

Lenalidomide

Bradycardia, edema


ARF Acute renal failure; CVA Cerebrovascular accident; DVT Deep vein thrombosis; SIAD Syndrome of inappropriate antidiuretic hormone; MI Myocardial ischemia; TIA Transient ischemic attack

Chemotherapy regimens are often used as combination therapies to target different pathways in the neoplastic process. Each medication’s metabolism, half-life, and excretion pathway influence its toxicity profile. Further, toxicities may contribute to or exacerbate coexistent comorbidities and polypharmacy exacerbates these issues [18]. For instance, cardiotoxicity and cardiac injury are common adverse effects of antimetabolites such as doxorubicin. Thus, preoperative cardiac evaluation must be enforced. Pulmonary toxicity is common with bleomycin, methotrexate, and mitomycin C therapy resulting in early plain radiological findings of fine, reticular bibasilar infiltrates. Clinically, these findings may affect anesthesia delivery and hemodynamic response. Methotrexate and azathioprine have been linked to hepatic fibrosis and intrahepatic cholestasis, respectively [18]. Renal toxicity is common among platinum compounds, methotrexate, and streptozocin but can be avoided with adequate hydration and avoidance of simultaneous use of renal toxic compounds such as aminoglycoside antibiotics. Nitrogen mustards such as cyclophosphamide and ifosfamide often cause hemorrhagic cystitis. This is a self-limiting effect and cease after discontinuation of therapy. Treatment with mesna for hemorrhagic cystitis is useful, but definite treatment may require cystoscopy-assisted hemostasis through electrocautery or sclerosis with formalin [18]. Early manifestations of neurotoxicity include neuropathy and hyporeflexia. However, neurotoxicity may progress to SIADH, tinnitus, hearing loss, autonomic instability, cranial nerve palsies, and emesis related to alkaloids, antimetabolites, or platinum agents. Emesis is a common ailment and should be readily anticipated and treated prior to chemotherapy induction.

Myelosuppression is dose limiting in many antineoplastic regimens, yielding reduced production of blood cell precursors approximately 2 weeks after induction. This cytopenia is commonly transient, lasting several days, but marked by neutropenia and absolute neutrophil count (ANC) <1000 cell/mm3 [19]. Neutropenic patients lack the classic signs of infection (erythema and elevated cell counts) and most frequently present with fever as the sole complaint in 25–40 % of patients. Hypotension, weakness, and confusion may also occur. The source of the fever is rarely found, warranting implementation of antimicrobial therapy per Infectious Diseases Society of America (IDSA) guidelines for neutropenic patients [20]. These guidelines recommend expanded broad-spectrum coverage for gram-negative and anaerobic organisms with piperacillin–tazobactam or a carbapenem [20]. Combination therapy with antipseudomonal cephalosporins plus metronidazole is also appropriate. Therapy should be continued until resolution of symptoms and objective signs of improved neutrophil count (ANC >500 cell/mm3) [20]. Antifungal coverage should be started with amphotericin B in the setting of continued fever, neutropenia, and clinical deterioration or lack of improvement after 5 days of adequate antimicrobial coverage. Addition of colony-stimulating factor (CSF) to boost immune response can be considered in septic patients or if ANC falls below 100 cell/mm3 [20].

Achieving the ideal chemotherapeutic regimen includes a balance between maximizing tumor suppression and local control, and minimizing toxicity and immunosuppression. Surgeons must be able to recognize and properly evaluate antineoplastic drug toxicities in preoperative planning to reduce surgical complications. Appropriate treatment for neutropenia should focus on antimicrobial therapy and boosting the immune system prior to surgical intervention.


Biologic Therapy for Rheumatic and Autoimmune Disease


The use of targeted biologic therapy has emerged as a suitable approach for patients with rheumatic and autoimmune diseases. The mechanism of action of these agents includes reduction of chronic inflammatory changes by targeting cytokines, inflammatory cells, and costimulation molecules [21]. Considering risk and benefits of primary disease flare with serious infections from surgical complications is an important distinction.

Cytokines such as tumor necrosis factor (TNF-α), Interleukin-1 (IL-1), and Interleukin-6 (IL-6) have major roles in endothelial cell activation, promoting inflammation, coagulation, wound healing, and hepatic synthesis of acute phase reactants [21]. IL-6 is also known for B-cell activation and antibody proliferation. Additionally, cluster of identification (CD) molecules are cell surface molecules involved in activation of B and T lymphocytes particularly CD 20, 22, and 28 [22]. Drugs that target these pathways are of particular importance for the management of chronic inflammatory and autoimmune conditions. Table 22.4 includes a list of currently FDA-approved agents and indications for use.


Table 22.4
Biologic therapy and indications for use























































Target

Agent

Indications and uses

TNF-a

Etarnecept

RA, PsA, JIA, plaque psoriasis

Infliximab

RA, CD, UC, plaque psoriasis, PsA, Takayasu’s vasculitis

Adalimumab

RA, CD, UC, AS, PsA

Golimumab

RA, PsA, AS, UC

Certolizumab

RA, AS, Crohn’s disease, PsA

IL-1

Anakinra, rilonacept, Canakinumab

RA, CAPS

IL-6

Tocilizumab

RA, JIA

INF-B1α

Avonex

Multiple sclerosis

CD-20

Rituximab

RA, MPA, pemphigus vulgaris, leukemia, lymphoma, Wegener’s granulomatosis

Ofatumumab

RA, leukemia

CD-22

Epratuzumab

SLE, Sjögren’s syndrome

CD-28

Abatacept

RA, JIA


AS Ankylosing spondylitis; CAPS Cryopyrin-associated periodic syndrome; CD Crohn’s disease; INF Interferon; ILK Interleukin; JIA Juvenile idiopathic arthritis; MPA Microscopic polyangiitis; PsA Psoriatic arthritis; RA Rheumatoid arthritis; SLE Systemic lupus erythematosus; TNF Tumor necrosis factor; UC Ulcerative colitis

The initiation of biologic treatment requires screening for previous or current tuberculosis infection, serological evidence of viral hepatitis, and previous or current malignancy. This is important as therapy with biologics can induce reactivation of latent infections [23]. Risks of myelosuppression and decreased wound healing are also prominent [24]. This infection risk is heightened by the inherent risk of autoimmune diseases. For instance, RA patient have been found to have a 13-fold increase in infection risk when compared to healthy controls in the absence of immunosuppressive regimen [25]. Interestingly, there is a lack of recommendations regarding continued use of biologics in the perioperative period but most supports cessation of therapy should be practiced when possible.

Anti-TNF-α agents have emerged as effective therapies for inflammatory bowel disease. Rituximab, originally used in the treatment of lymphoma, is a monoclonal antibody against CD 20 cells, which represses a subset of B cells via apoptosis [24]. It is approved for use in autoimmune disorders and lymphocytic malignancies but is also used off label for auto-autoimmune disorders (idiopathic thrombocytopenic purpura), desensitization of ABO blood groups in transplant incompatible patients, and dermatologic disorders [24]. IL-1 inhibitors, such as Anakinra, have few clinical indications and a short half requiring daily injections and common development of skin tissue infections at site of injection. Surgical implications of biologic are discussed separately in this chapter.


Transplant


The total number of transplants performed in the USA in 2011 reached over 29,000 [26]. Many transplant recipients are experiencing longer survival in part due to better understanding of pathophysiology involved in rejection, improvements in immunosuppressive therapy, and more efficient immunosuppressive agents combination strategies. Post-transplant immunosuppression is typically achieved with glucocorticoids, calcineurin inhibitors, antiproliferative, and/or antimetabolic drugs [18]. Surgeons must have an understanding of these immunosuppressive therapies for appropriate management of acute and elective interventions (Table 22.5).


Table 22.5
Transplant immunosuppressive agents, indications, and common adverse effects






































Class

Agent

Indication

Toxicity

Corticosteroids

Prednisone, methylprednisolone, hydrocortisone

Maintenance for most transplants, high doses for acute rejection, preoperative “stress dose”

Early: Hyperglycemia, avascular necrosis of the hip, serious infections

Late: Impaired wound healing, muscle wasting, osteoporosis, Cushing’s syndrome, adrenal insufficiency during stress

Calcineurin inhibitors (CNI)

Tacrolimus and cyclosporine

Maintenance therapy for cardiac, renal, and hepatic transplants

Nephrotoxicity, CNS effects (seizure, coma, tremors, and headache), hyperglycemia, hypomagnesemia, hypertension, dyslipidemia, ischemic events (CVA, MI)

Antimetabolite

Sirolimus and Everolimus

Maintenance, preferred in renal transplants over CNI

Anemia, leukopenia, thrombocytopenia, impaired wound healing, hypo/hyperkalemia

Antiproliferative

Azathioprine

Chemotherapy, transplant maintenance immunosuppression

Myelosuppression, hepatotoxicity, pancreatitis, depressed neuromuscular blockade from anesthetics

Mycophenolic acid, mycophenolate mofetil

Transplant maintenance immunosuppression

Leukopenia, CMV sepsis

Glucocorticoids are the oldest medications used in transplant immunosuppression. Immunosuppression is achieved by reducing T-cell proliferation, inflammatory cytokines, vasodilatory molecules, tissue permeability, and preventing monocyte migration [27]. They exert their role by binding to intracellular receptors and modulating cellular transcription [27]. Initial administration results in neutrophilic leukocytosis, eosinophilia, and lymphocytopenia. Interestingly, while there is a reduction in total number of B cells, immunoglobulin production is preserved [28]. Additionally, T-cell apoptosis results in impaired cell-mediated immunity [27]. The side effect profile of corticosteroid depends on amount, steroid potency, mineralocorticoid profile, and the length of treatment. Major side effects include hypertension, cardiovascular disease, hypokalemia, adrenal insufficiency, diabetes, visceral perforation, and higher risk of infection. Surgical complications and perioperative management in the setting of steroid use deserve further consideration and will be addressed later in this chapter.

Calcineurin inhibitors (CNI) are the most commonly used agents post-transplantation and include tacrolimus and cyclosporine (CsA). These agents substantially decreased organ rejection and revolutionized transplant immunosuppression when first introduced. Their mechanism of action involves blockage of NFAT (nuclear factor of activated T cells) transcription reducing cytokine release from T cells [29]. Dosage requires close monitoring due to narrow therapeutic range contingent on type of organ transplant. Oral administration requires GI absorption, P450 hepatic metabolism and bile excretion [29]. Thus, any metabolic (P450 inducer/inhibitor), physiological (alteration of GI flora, diarrhea, inflammation), or mechanical process can affect CNI metabolism. CNIs have nephrotoxic, neurotoxic, metabolic, and endocrine side effects [29]. The nephrotoxic effects are particularly important after renal transplant often requiring dosing reduction or selection of different agents. Avoiding concomitant use of nephrotoxic drugs such aminoglycosides is strongly encouraged. Neurotoxicity presents as seizure, confusion, coma, tremors, or headache [30]. Multiple electrolyte and metabolic derangements also occur including hyperglycemia, hypomagnesemia, and hypercholesterolemia [31]. Tacrolimus-induced hyperglycemia occurs by establishing insulin resistance that is further exacerbated by surgical stress. Postoperative treatment with a sliding insulin regimen may be required in a patient with otherwise adequate intrinsic glucose control [31]. Higher incidence of ischemic events (CVA and MI) complicates preoperative planning and places these patients at higher risk [31].

Antiproliferative and antimetabolic drugs include azathioprine, mycophenolic acid, mycophenolate mofetil (MMF), sirolimus, and everolimus. Azathioprine, often used in chemotherapy, interferes with DNA synthesis by incorporating false nucleotides (6-thio-GTP) into replicating strand of rapidly proliferative cells such as B and T lymphocytes [32]. Main adverse effects of azathioprine include hepatotoxity, pancreatitis, and dose-limiting myelosuppression [32]. MMF and mycophenolic acid inhibit purine synthesis and have similar preference for fast replicating cells, thus sharing side effects of azathioprine. Sirolimus (Rapamycin) and everolimus, similar to tacrolimus, reduce lymphocytic activation by inhibition of mTOR (mechanistic Target of Rapamycin) and IL-2 signaling [33]. Sirolimus has a preferred renal profile over CNI and is often used as maintenance in renal transplant. However, these agents induce severe leukopenia and thrombocytopenia. Sirolimus also decreases wound healing resulting in dehiscence and incisional hernias particularly in obese patients with BMI >32 kg/m2 [33]. In this group, it is beneficial to switch to CNI therapy for elective surgeries for two to four months prior and restarting 6-week postoperatively to improve wound healing.

Mucosal injury and ulceration are common ailments among transplant immunosuppressive therapy. Diarrhea and GI side effects are more common with tacrolimus, CsA, and MMF in a dose-dependent fashion [34]. A proposed mechanism for MMF-associated diarrhea includes inhibition of cryptcell division and loss of duodenum villous structure [34]. Ulceration of the GI tract is multifactorial, but has been associated with utilization of NSAIDs, surgery stress, and impaired cytoprotection of mucosal defenses from decreased cell turnover [34]. GI bleeding, when it occurs, is often secondary to undiscovered ulceration. Lung transplant patients may acquire giant gastric ulcers, defined as ulcers with diameter >3 cm, resulting in high mortality secondary to bleeding [34]. Identifying patients with risk factors for GI ulcers is difficult and low threshold for routine endoscopy with biopsy in symptomatic patients is endorsed. Standard practice should include ulcer prophylaxis with PPI or H2 blockers. Perforations can occur throughout the GI tract secondary to steroids, diverticular disease, and concomitant NSAIDs use. Transplant recipients are also at increased risk for biliary tract disease from calculi influenced by cell turnover, resulting in emergent cholecystectomies with high mortality rates. In renal transplant recipients, cyclosporine has resulted in reduced bile flow and increased incidence of cholelithiasis [34]. Therefore, elective surgery for eradication of biliary pathology and ultrasound screening should be practiced during the post-transplantation period to improve outcomes. Acute pancreatitis, although uncommon, carries increased mortality rates of 64 % in liver transplants and 100 % in renal transplant patients [33]. Risk factors for acute pancreatitis include immunosuppressive agents, CMV, HBV, hypercalcemia, alcohol, cholelithiasis, and malignancy [34]. Follow-up with CT at regular intervals to identify unusual inflammation, pseudocyst formation, edema, or necrosis is beneficial. Treatment for acute pancreatitis must focus on aggressive intravenous fluid resuscitation, fasting, identification, and cessation of inciting agent followed by surgery when clinically indicated [33].

Infectious complications are of particular importance in transplant patients due to the nature of chronic immunosuppression. As previously addressed, atypical presentation of infection is common among this group of patients and the clinician must be alert and cautious during preoperative planning. Lung transplant recipients experience invasive early and late fungal infections with coccidiomycosis and aspergillus, respectively [35]. Prophylactic treatment with fluconazole is recommended for the former [36]. Cutaneous and anogenital lesions should raise concern for HPV infections and marked immunosuppression requiring biopsy and surveillance [37]. Finally, CMV infection is the most common infection among post-transplant recipients and is associated with allograft rejection, EBV post-transplantation proliferative disorder (PTLD), and high rate of mortality in the first 6 months [38].

Transplant patients represent a complex mixture of pathologies requiring multidisciplinary collaboration between physicians (transplant, infectious diseases, PCP, and surgeons) for appropriate management and risk reduction. When appropriate, termination or substitution of immunosuppressive regimen improves surgical outcomes.



Surgical Diseases



Gastrointestinal Diseases


The gastrointestinal tract is susceptible to disease from immunosuppression for multiple factors. First, antineoplastic agents are cytotoxic and affect GI cell turnover. Drug immunomodulation modifies components of innate immunity such as barrier protection, neutrophil migration, and resident dendritic cells reducing host defenses. Antibiotic regiments often used in immunocompromised patients alter the inhabitant flora selecting for virulent organisms. Finally, tissue inflammation and ischemia affect the GI tract requiring appropriate management to avoid exacerbation.


Typhlitis/Neutropenic Enterocolitis


Typhlitis describes a syndrome of gastrointestinal inflammatory conditions characterized by segmental ulceration and necrotizing inflammation of the terminal ileum, cecum, and/or ascending colon in neutropenic patients [39]. This syndrome, also referred to as neutropenic enterocolitis, ileocecal syndrome, cecitis, necrotizing enterocolitis, and agranulocytic colitis, was initially described as necrotizing cecal enteropathy in children undergoing chemotherapy for leukemia or lymphoma [40]. Typhlitis is the most common gastrointestinal infection involving neutropenic patients with incidence reaching 5.3 % in patients undergoing systemic chemotherapy and in aplastic anemia [41]. Pathogenesis of this syndrome is related to cytotoxic mucosal injury during chemotherapy that affects the immunological surface barriers resulting in bacterial and toxin translocation into the submucosa with corresponding inflammation, ulceration, and transmural necrosis [42]. This is complicated by multiple factors including neutropenia, poor host defenses against gastrointestinal flora, and superimposed infection [43]. Regional preference for the cecum is related to decreased vascularity and increased concentration of lymphatic tissue.

Clinical presentation in a subset of patients includes the triad of right lower quadrant abdominal pain, fever and neutropenia along with non-specific symptoms of nausea, vomiting, abdominal distention, and watery or bloody diarrhea [44]. Physical examination is often non-specific with abdominal distention and pain localization to the right lower quadrant in 40–69 % of patients [45]. An acute abdomen is rarely the presenting symptom and high index of suspicion should be practiced to prevent complications in neutropenic patients presenting with abdominal pain. As clinical presentation is often vague, imaging studies such as computed tomography (CT) and ultrasound (US) increase the likelihood of diagnosis.

Transmural bowel wall thickening >10 mm has been associated with increased mortality and some authors have proposed thickening >4 mm as a diagnostic criteria in the setting of neutropenia, fever, and abdominal pain [46]. CT delivers the best evaluation of bowel wall thickening. CT with oral and intravenous contrast provides a detailed picture of bowel integrity, pneumatosis intestinalis, free air, and surrounding edema or hemorrhage [47]. Ultrasound can expedite diagnosis of typhlitis by demonstrating a doughnut-like, hypoechoic fluid filled intestinal lumen separated by a thickened hyperechoic mucosal wall [48]. Early inflammatory changes identified by ultrasound can expedite medical management and reduce operative indications especially in the pediatric population where radiation exposure is of concern. Plain radiography rarely provides additional information. The mortality rates of neutropenic patients necessitating surgery have been reported as high as 57 %; thus, medical management is often the recommended initial approach focusing on improving neutropenia, decreasing inflammation, and antibiotic therapy [43]. Further medical management includes bowel rest, nasogastric tube decompression, fluid resuscitation, and antipyretic therapy. Bone marrow stimulation should be considered per Infectious Disease Society of America guidelines for all neutropenic patients who fail to improve on initial broad-spectrum antibiotics, or have worsening neutropenia [49]. Surgical intervention is considered as a last resort therapy.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Nov 18, 2017 | Posted by in Uncategorized | Comments Off on The Immunosuppressed Patient

Full access? Get Clinical Tree

Get Clinical Tree app for offline access