Pancreas Transplantation Alone (PTA) and Islet Cell Transplantation

Number: 0601

Table Of Contents

Policy
Applicable CPT / HCPCS / ICD-10 Codes
Background
References


Policy

Scope of Policy

This Clinical Policy Bulletin addresses pancreas transplantation alone (PTA) and islet cell transplantation for commercial medical plans. For Medicare criteria, see Medicare Part B Criteria.

Note: Requires Precertification: 

Precertification of donislecel-jujn (Lantidra) suspension is required of all Aetna participating providers and members in applicable plan designs. For precertification of Lantidra, contact National Medical Excellence (NME) at 877-212-8811.  

  1. Medical Necessity

    Aetna considers pancreas transplantation alone (PTA) and islet cell transplantation medically necessary when criteria are met (unless otherwise stated): 

    1. Pancreas Transplantation Alone (PTA)

      1. PTA without kidney transplant for members who meet the transplanting institution's selection criteria. In the absence of an institution's selection criteria, Aetna considers PTA without kidney transplant medically necessary when all of the following general and disease specific criteria are met:

        1. General Criteria:
          1. Absence of ongoing or recurrent active infections that are not effectively treated; and
          2. Controlled HIV/AIDS infection, defined as:

            1. CD4 count greater than 200 cells/mm3 for more than 6 months; and
            2. HIV-1 RNA (viral load) undetectable; and
            3. No other complications from AIDS, such as opportunistic infection (e.g., aspergillus, coccidioidomycosis, resistant fungal infections, tuberculosis) or neoplasm (e.g., Kaposi's sarcoma, non-Hodgkin's lymphoma); and
            4. On stable anti-viral therapy more than 3 months; and
          3. Documentation of compliance with medical management; and
          4. Member has adequate cardiac status (e.g., no angiographic evidence of significant coronary artery disease, ejection fraction greater than or equal to 40%, no myocardial infarction in last 6 months, negative stress test); and
          5. Member has satisfactory kidney function (creatinine clearance greater than 40 ml/min); and
          6. No malignancy (except for non-melanomatous skin cancers or low-grade prostate cancer) or malignancy has been completely resected OR (upon medical review) malignancy has been adequately treated such that the risk of recurrence is small;
        2. Disease Specific Criteria:
          1. Member has a history of labile (brittle) insulin-dependent diabetes mellitus (IDDM); and
          2. Member has recurrent, acute and severe metabolic and potentially life-threatening complications requiring medical attention, as documented by chart notes, frequent emergency room visits and/or hospitalizations. They may include:

            1. Hyperglycemia; or
            2. Hypoglycemia; or
            3. Hypoglycemic unawareness associated with high risk of injury; or
            4. Ketoacidosis; and
          3. Member has consistent failure of exogenous insulin-based management, defined as inability to achieve sufficient glycemic control (HbA1c of greater than 8.0) or recurrent hypoglycemic unawareness, despite aggressive conventional therapy including all of the following:

            1. Adjusting frequencies and amounts of insulin injected; and
            2. Measuring multiple blood glucose levels on a daily basis; and
            3. Modifying diet and exercise; and
            4. Monitoring HgbA1c levels;
      2. Pancreas retransplantation after a failed primary pancreas transplant when member meets the selection criteria stated above;
      3. Pancreas retransplantation after 2 or more prior failed pancreas transplants may be considered medically necessary upon individual case review;
      4. Contraindications:
        1. Absolute Contraindications:

          PTA is considered not medically necessary in members with prohibitive cardiovascular risk because the risks of PTA exceed the benefits. Examples of prohibitive cardiovascular risk include, but are not limited to:

          1. Angiographic evidence of significant non-correctable coronary artery disease; or
          2. Ejection fraction less than 35%; or
          3. Myocardial infarction within last 6 months; or
          4. Refractory uncontrolled hypertension;
        2. Relative Contraindications:

          Aetna considers pancreas transplant medically necessary for members with the following relative contraindications to pancreas transplant only if the requesting physician documents that these relative contraindications were considered, and has determined that the benefits of pancreas transplant outweigh the risks in these members. Relative contraindications to PTA include the following:

          1. Ejection fraction 35 % to 40 %; or
          2. Severe peripheral vascular disease; or
          3. Unstable alcohol or substance use disorder (exception to this contraindication will be allowed with supporting documentation (within 4 weeks) demonstrating 3 months of stability from treating addiction medical professional or psychiatrist);
      5. Partial pancreas transplant from a living donor is considered an acceptable alternative to cadaveric transplant for persons who meet medical necessity criteria for pancreas transplant;

    2. Islet Cell Transplantation

      1. Allotransplantation (i.e., transplantation of islet cells from a donor; also known as allogeneic pancreatic islet cellular therapy) with FDA-approved donislecel-jujn (Lantidra) suspension for hepatic portal vein infusion when all of the following criteria are met:

        1. Criteria for Initial Approval
          1. Member is 18 years of age or older; and
          2. Member has type 1 diabetes and is unable to approach target hemoglobin A1C (HbA1c) due to current repeated episodes of severe hypoglycemia despite intensive diabetes management and education; and
          3. Lantidra will be used in conjunction with concomitant immunosuppression therapy (e.g., mycophenolate mofetil, sirolimus, tacrolimus); and
          4. Member has not received a renal transplant; and
          5. Administration has not exceeded a maximum of 3 infusions (transplants);
        2. Continuation of Therapy

          Lantidra infusion (transplant) may be repeated when member meets all initial approval criteria and has not achieved independence from exogenous insulin within one year of infusion or within one year after losing independence from exogenous insulin after a previous infusion. (Note: There are no data regarding the effectiveness or safety for more than 3 infusions).

      2. Autotransplantation (i.e., transplantation of the member's own islet cells) for members undergoing near-total or total pancreatic resection for severe, refractory chronic pancreatitis.

  2. Experimental and Investigational

    Aetna considers any of the following procedures experimental and investigational because their safety and effectiveness have not been established in the peer-reviewed published medical literature (not an all-inclusive list):

    1. Ex-situ machine pancreas perfusion (in hypothermic or normothermic settings) for pancreas transplantation; or
    2. Pancreatic islet xenotransplantation; or
    3. PTA for increased longevity; or
    4. PTA primarily indicated for life-style issues, i.e., a desire to no longer take insulin.

Dosage and Administration

Lantidra dosage form is a cellular suspension of allogeneic pancreatic islets (islets of Langerhans) in buffered transplant media. Dosage strength depends on the total number of islets packaged for infusion, which is reported on the container label and associated documents.

The recommended minimum dose is 5,000 equivalent islet number (EIN) per kg person's body weight for initial infusion (transplant) and 4,500 EIN/kg for subsequent infusions (same recipient). The maximum dose per infusion is dictated by the estimated tissue volume, which should not exceed 10 cc per infusion, and the total EIN present in the infusion bag (up to a maximum of 1 x 106 EIN per bag).

A second infusion may be performed if the individual does not achieve independence from exogenous insulin within one year of infusion or within one year after losing independence from exogenous insulin after a previous infusion.

A third infusion may be performed using the same criteria as for the second infusion. There are no data regarding the effectiveness or safety for persons receiving more than three infusions.

Immunosuppression is to be continued permanently to prevent islet graft rejection.

Source: CellTrans, 2023


Table:

CPT Codes / HCPCS Codes / ICD-10 Codes

Code Code Description

CPT codes covered if selection criteria are met:

0584T Islet cell transplant, includes portal vein catheterization and infusion, including all imaging, including guidance, and radiological supervision and interpretation, when performed; percutaneous
0585T      laparoscopic
0586T      open
48160 Pancreatectomy, total or subtotal, with autologous transplantation of pancreas or pancreatic islet cells
48550 Donor pancreatectomy (including cold preservation), with or without duodenal segment for transplantation
48551 Backbench standard preparation of cadaver donor pancreas allograft prior to transplantation, including dissection of allograft from surrounding soft tissues, splenectomy, duodenotomy, ligation of bile duct, ligation of mesenteric vessels, and Y-graft arterial anastamoses from iliac artery to superior mesenteric artery and to splenic artery
48552 Backbench reconstruction of cadaver donor pancreas allograft prior to transplantation, venous anastamosis, each
48554 Transplantation of pancreatic allograft
48556 Removal of transplanted pancreatic allograft

CPT codes not covered for indications listed in the CPB:

Ex-situ machine pancreas perfusion- no specific code

Other CPT codes related to the CPB:

36481 Percutaneous portal vein catheterization by any method [Hepatic portal vein infusion]
80069 Renal function panel
82947 Glucose; quantitative, blood (except reagent strip)
82948     blood, reagent strip
82950     post glucose dose (includes glucose)
82962 Glucose, blood by glucose monitoring device(s) cleared by the FDA specifically for home use

HCPCS codes covered for indications listed in the CPB:

Donislecel-jujn (Lantidra)-no specific code
S2102 Islet cell tissue transplant from pancreas; allogeneic

HCPCS codes not covered for indications listed in the CPB:

G0341 Percutaneous islet cell transplant, includes portal vein catheterization and infusion
G0342 Laparoscopy for islet cell transplant, includes portal vein catheterization and infusion
G0343 Laparotomy for islet cell transplant, includes portal vein catheterization and infusion

ICD-10 codes covered if selection criteria are met:

E08.649 Diabetes mellitus due to underlying condition with hypoglycemia without coma
E10.10 - E10.9 Type 1 diabetes mellitus
E11.00 - E11.9 Type 2 diabetes mellitus
E15 Nondiabetic hypoglycemic coma
E16.0 - H16.2 Hypoglycemia
E79.0 Hyperuricemia without signs of inflammatory arthritis and tophaceous disease
E87.20, E87.21, E87.22, E87.29 Acidosis
E89.1 Postprocedural hypoinsulinemia
K86.0 - K86.1 Chronic pancreatitis
R78.71 Abnormal lead level in blood
R78.79 Finding of abnormal level of heavy metals in blood
R78.89 Finding of other specified substances, not normally found in blood
R79.0 Abnormal level of blood mineral
R79.9 Abnormal finding of blood chemistry, unspecified
Z79.4 Long term (current) use of insulin
Z90.410 Acquired total absence of pancreas
Z90.411 Acquired partial absence of pancreas

ICD-10 codes absolute contraindication for this CPB:

I10 Essential (primary) hypertension
I15.0 - I15.9 Secondary hypertension
I16.0 - I16.9 Hypertensive crisis
I21.01 - I21.A9 Acute myocardial infarction
I22.0 - I22.9 Subsequent ST elevation (STEMI) and non-ST elevation (NSTEMI) myocardial infarction
I25.10 - I25.119 Atherosclerotic heart disease of native coronary artery

ICD-10 codes relative contraindication for this CPB:

F10.10 - F19.99 Mental and behavioral disorders due to psychoactive substance use
I73.00 - I73.9 Other peripheral vascular diseases

Background

Exogenous insulin is effective therapy for most diabetics.  Intensive insulin therapy with multiple daily injections or with a constant infusion pump has been shown to be an effective method of maintaining blood glucose and hemoglobin A1c at near normal levels.  Despite maximal medical therapy, a rare (less than 1%) number of non-uremic patients with insulin-dependent diabetes mellitus (IDDM) may experience frequent and unpredictable occurrences of severe hyperglycemia, hypoglycemia, ketoacidosis, and hypoglycemic unawareness, thus labeling them as brittle, labile and unstable.  This subgroup tends to have a life that is constantly disrupted by these disabling and life-threatening events, which cause a considerable burden on social and family resources due to multiple emergency room visits and/or hospital admissions.  Fortunately, in this small subset of patients, there is sufficient evidence in the medical literature to support performance of pancreas transplantation alone (PTA).

Pancreas Transplantation Alone (PTA)

Initially, the reported results of solitary pancreatic transplantation in non-uremic diabetic patients were less favorable than simultaneous pancreas-kidney (SPK) transplantation or pancreas after kidney (PAK) transplantation because of high rates of rejection and difficulties in the diagnosis and treatment of these rejection episodes.  Because there was no marker for pancreas rejection with sensitivity similar to serum creatinine for kidney transplant rejection, detection and treatment of PTA rejection was often delayed.  Subsequently, outcomes of PTA have substantially improved due to technical refinements of the procedure, the introduction of new immunosuppressive regimens, and better selection of transplant candidates.  Unlike SPK and PAK, there is no adequate evidence that PTA can prevent or retard the development and/or progression of the long-term complications of diabetes; nor is there evidence that pancreas transplantation can prolong the life of patients with diabetes mellitus

Pancreas transplantation alone is contraindicated in patients with clinically significant cardiovascular disease.  Because of the high prevalence of microvascular disease, especially in the coronary arteries in patients with diabetes, it is especially important to evaluate patients for these risks before PTA surgery.  Indeed, one of the most frequent causes of pancreas transplant failure is death from myocardial infarction.  Therefore, accepted guidelines state that any substantial coronary artery disease needs to be corrected before transplantation is undertaken.

A history of coronary revascularization is no longer considered an absolute contraindication.  Nevertheless, coronary event rates remain greater among pancreas transplant recipients who have undergone coronary revascularization, than among individuals without pre-transplantation coronary artery disease.  The literature states that severe peripheral vascular disease is a relative contraindication to PTA since iliac atherosclerosis can complicate the technical procedure.  Other relative contraindications to pancreas transplantation include obesity, substance abuse, poorly controlled psychiatric illnesses, noncompliance, and any recent malignancy.

Although segmental grafts (consisting of only the pancreatic body and tail) were once common, the entire pancreas and its associated duodenal segment are now almost always transplanted (unless a living donor is used).  This advance provides more insulin-secreting cells.  The allograft is positioned laterally in the lower abdomen.  Vascular anastomoses in a pancreas transplant are the donor splenic and superior mesenteric artery and portal vein to the recipient iliac artery and vein, respectively.  This provides systemic rather than portal delivery of insulin with a resulting baseline fasting hyper-insulinemia.  The incidence of post-transplant graft thrombosis has been greatly reduced using this method.  Ligation or obliteration of the pancreatic duct was once commonly practiced, but these techniques have also been abandoned.  Instead, the pancreatic exocrine secretions are drained internally into either the small intestine or the bladder.  In general, immunosuppression is the same as that used for patients with kidney transplants.  Induction therapy and rejection treatment usually involves use of ALG or OKT3.  All in all, with excellent HLA matching, a graft survival rate of 80 %, comparable with the overall success rate of combined kidney-pancreas transplantation, can be achieved.

Indications for Pancreas Transplantation

The American Diabetes Association (2003) has developed established indications for pancreas transplantation: "In the absence of indications for kidney transplantation, pancreas transplantation should only be considered a therapy in patients who exhibit these 3 criteria:

  1. a history of frequent, acute, and severe metabolic complications (hypoglycemia, hyperglycemia, ketoacidosis) requiring medical attention;
  2. clinical and emotional problems with exogenous insulin therapy that are so severe as to be incapacitating; and
  3. consistent failure of insulin-based management to prevent acute complications."

Islet Cell Transplantation

Autotransplantation

Islet cell autotransplantation, also known as an autologous islet cell transplantation, is the infusion of a patient's own pancreatic islet cells into the portal vein of the liver. 

Autologous islet cell transplantation is an alternative for persons undergoing total pancreatectomy for severe, refractory chronic pancreatitis.  Near total or total pancreatic resection can alleviate pain in patients with severe chronic resection.  Autologous islet cell transplantation can preserve islet cell function in patients undergoing this procedure.  The islet cell transplantation procedure involves the infusion of islet cells into the liver by portal embolization, where the cells function as a free graft.  The liver's dual vascular supply allows embolization of isolated pancreatic islets by cannulating the umbilical vein, a tributary of the mesenteric venous system, or by transcutaneous, transhepatic cannulation of the portal vein itself.  The terminal portal venule can be occluded without infarcting the transplant site.

Rodriguez Rilo and associates (2003) reported on the results of autologous islet cell transplantation in a consecutive series of patients from one center who received total or near-total pancreatic resection for severe, refractory chronic pancreatitis.  From February 2000 to February 2003, a total of 22 patients, whose median age was 38 years, underwent pancreatectomy and autologous islet cell transplantation.  Sixty-eight percent of the patients had either a minor or major complication.  Major complications included acute respiratory distress syndrome (n = 2), intra-abdominal abscess (n = 1), and pulmonary embolism (n = 1).  All patients demonstrated C-peptide and insulin production indicating graft function.  Post-operatively, 41 % of subjects were insulin independent, and 27 % required less than 10 units of insulin per day, and the remaining 7 patients require between 15 and 40 units of insulin per day.  All patients had pre-operative pain and had been taking opioid analgesics; 82 % no longer required analgesics post-operatively.  The investigators concluded that pancreatectomy with autologous islet cell transplantation can alleviate pain for patients with chronic pancreatitis and preserve endocrine function.

Jie et al (2005) reported on outcomes of one center’s experience with 137 patients undergoing autologous islet cell transplantation for pancreatectomy since 1997.  Follow-up data was available in 120 patients; 63 % of the patients had complete relief from pain, 22 % experienced partial relief, and 15 % were unchanged.  Of patients with complete pancreatectomy since 1995 (n = 73), all but 1 pediatric patient (n = 22) transplanted with less than or equal to 2000 IEQ/kg islets required insulin post-pancreatectomy.  Of patients receiving more than 2000 IEQ/kg islets (n = 51), 47 % were completely insulin independent while 25 % were intermittently insulin-treated.  The investigators concluded that autologous islet cell transplantation should be considered in patients undergoing primary pancreatic resection for the treatment of refractory pain associated with small duct chronic pancreatitis.

Clayton and colleagues (2003) reported on a single center’s experience with autologous islet cell transplantation for total or partial pancreatectomy from September 1994 to July 2001.  Forty patients had been transplanted, with follow-up times range from 6 months to 7 years.  At 2 years post transplant, 18 patients had a median hemoglobin A1c of 6.6 % (5.2 to 19.3 %), fasting C-peptide of 0.66 ng/mL (0.26 to 2.65 ng/ml), and required a median of 12 (0 to 45) units of insulin per day.  Five patients with 6-year follow-up data had a median hemoglobin A1c of 8 % (6.1 to 11.1 %), fasting C peptide of 1.68 ng/ml (0.9 to 2.78 ng/ml), and required a median of 43 (6 to 86) units of insulin per day.  The investigators reported that these data demonstrate that up to 6 years after autologous islet cell transplantation, the grafts continue to function, but that over the time period studied, the level of function appears to be decreasing.  The investigators reported that the majority of patients no longer required opiate analgesia.

Allotransplantation

Islet cell allotransplantation is the transplantation of islet cells from a genetically non-identical donor of the same species. This type of transplantation is also known as allogeneic pancreatic islet cellular therapy.

Allogeneic islet cell transplantation has been investigated as an alternative means of restoring normoglycemia, without the attendant morbidity of the whole-organ procedure.

Guidelines from the American Diabetes Association (Robertson et al, 2004) concluded that "islet cell transplantation is an experimental procedure, also requiring systemic immunosuppression, and should be performed only within the setting of controlled research studies."

An assessment prepared for the Ontario Ministry of Health and Long-Term Care (2003) concluded that "[t]he current evidence on the use of islet transplantation for non-uremic type 1 diabetic patients is limited since it is based on studies with weak methodological design .… The effect of islet transplantation on restoring hormonal responses to hypoglycemia is inconclusive. Islet transplantation in non-uremic type 1 diabetic patients with hypoglycemia unawareness or uncontrolled diabetes is an evolving procedure with promising preliminary, but inconclusive final results."

An assessment prepared by the Alberta Heritage Foundation for Medical Research (Guo et al, 2003) reached the following conclusions about islet cell transplantation for type 1 diabetes: "Limited evidence suggests that ITA [islet cell transplantation] is effective in controlling labile diabetes and protects against unrecognised hypoglycemia in highly selected patients in the short term.  The long-term effects of ITA on metabolic control remain to be proven.  Follow-up studies are needed to determine the duration of this metabolic effect in order to assess its potential for preventing or arresting the development of chronic diabetes complications in non-uremic type 1 diabetic patients with severe hypoglycemia.  Future research is required to improve measures for islet mass/function in order to appropriately evaluate the effects of the ITA procedure."

An assessment by the National Institute for Clinical Excellence (2003) states: "Current evidence on the safety and efficacy of pancreatic islet cell transplantation does not appear adequate to support the use of this procedure without special arrangements for consent and for audit or research."  The assessment explained that "the identified studies did not compare blood sugar control or risks of diabetic complications for the treatment options (injected insulin versus pancreatic islet cell transplantation)."  The assessment also stated that there was a lack of long-term follow-up data.

An assessment of islet cell transplantation for type 1 diabetes prepared for the Agency for Healthcare Quality and Research (AHRQ) reached the following conclusions: "Evidence on outcomes of islet transplant is limited by small patient numbers, short followup, and lack of standardized reporting.  (These issues are being addressed by the NIH funded Collaborative Islet Transplant Registry.)  Of 37 patients from 3 centers, 28 (76 %) maintained insulin independence at 1 year (published evidence); similarly, 50 to 90 % of 104 patients from four centers were insulin independent (supplemental evidence).  Serious adverse events, including portal vein thrombosis and hemorrhage, occur infrequently.  Data are lacking on long-term durability of the procedure, effects on diabetic complications, or long-term consequences of immunosuppression.  Evidence is insufficient for comparison with whole-organ pancreas transplant."

An assessment by the Institute for Health Economics (Guo et al, 2008) of islet cell transplantation for type 1 diabetes concluded that there was insufficient evidence to consider islet cell allotransplantation as standard care in patients with non-uremic type-1 diabetes with severe hypoglycaemia and uncontrolled diabetes.  The assessment stated a number of implications for research, including the development of more sensitive methods to predict and detect graft loss, and the need for studies that were larger, prospective and had longer follow-up periods.  Studies of single donors with standardized immunosuppressive regimens and studies of patients with and without renal dysfunction were also recommended.

In a pilot study, Mineo and colleagues (2008) attempted to induce recipient chimerism and graft tolerance in islet transplantation by donor CD34+hematopoietic stem cell (HSC) infusion.  A total of 6 patients with brittle type 1 diabetes mellitus received a single-donor allogeneic islet transplant (8611 +/- 2113 IEQ/kg) followed by high doses of donor HSC (4.3 +/- 1.9 x 10(6) HSC/kg), at days 5 and 11 post-transplant, without ablative conditioning.  An "Edmonton-like" immunosuppression was administered, with a single dose of infliximab added to induction.  Immunosuppression was weaned per protocol starting 12 months post-transplant.  After transplantation, glucose control significantly improved, with 3 recipients achieving insulin-independence for a short time (24 +/- 23 days).  No severe hypoglycemia or protocol-related adverse events occurred.  Graft function was maximal at 3 months then declined.  Two recipients rejected within 6 months due to low immunosuppressive trough levels, whereas 4 completed 1-year follow-up with functioning grafts.  Graft failure occurred within 4 months from weaning (478 +/- 25 days post-transplant).  Peripheral chimerism, as donor leukocytes, was maximal at 1-month (5.92 +/- 0.48 %), highly reduced at 1-year (0.20 +/- 0.08 %), and was undetectable at graft failure.  CD25+T-lymphocytes significantly decreased at 3 months, but partially recovered thereafter.  Combined islet and HSC allotransplantation using an "Edmonton-like" immunosuppression, without ablative conditioning, did not lead to stable chimerism and graft tolerance.

In a review on pancreas retransplants, Humar et al (2000) concluded that pancreas retransplants can be performed with a minimal increase in surgical complications.  However, graft survival after retransplants is slightly inferior to that after primary transplants, probably for both immunological and non-immunological reasons.  Retransplants can be offered to suitable candidates, but they may require more aggressive monitoring for rejection.

Patients with type 1 diabetes who are appropriate candidates for a pancreas transplantation may be simultaneously evaluated for suitable living segmental pancreas donors (Barr et al, 2006).  Potential donors may undergo either segmental pancreas donation alone (for non-uremic or post-uremic recipients) or simultaneous segmental pancreas and unilateral kidney donation (for uremic recipients).  Once identified, potential donors will be subject to a thorough medical, metabolic and psychosocial screening.  ABO and HLA cross-match compatibility is preferred but not manditory (Barr et al, 2006).  A segmental donor pancreatectomy can also be applied for islet isolation and allotransplantation.  Donor segmental pancreatectomy (tail) can be done open or laparoscopically.

The largest reported experience with living donor pancreas transplants has been reported from the University of Minnesota (Barr et al, 2006).  At the University of Minnesota, there have been 130 live donor pancreas transplants performed between 1977 and 2005.  The distribution of these transplants was as follows: 40 % PTA; 25 % pancreas after kidney (PAK); and 35 % simultaneous live donor pancreas and kidney transplants (SPK).  There are 20 PTA and PAK live donor grafts functioning between 10 and 30 yeaars following transplantation.  There are 3 living donor SPK transplants with  function greater than 10 years.  There have also been 2 live donor islet cell transplants after kidney transplantation early in the center experience.

Other centers have also reported their experience with living donor pancreas transplantation.  At the University of Illinois, Chicago, 9 living-donor simultaneous kidney and segmental pancreas bladder-drained transplants were performed between 1997 and 2004 (Barr et al, 2006).  Eight out of 9 pancreas grafts and all the kidney grafts are reported to be working for 1 to 8 years following transplantation.  There was no report of a donor death.

As of 2005, there had been 5 live donor segmental pancreatectomies performed in Japan, 1 case of live donor islet cell transplantation in Japan, and 2 live donor segmental pancreatectomies performed in Korea (Barr et al, 2006).

Tan et al (2008) evaluated the safety and effectiveness of simultaneous islet and kidney transplantation in patients with type 1 diabetes and end-stage renal disease using a glucocorticoid-free immunosuppressive regimen with alemtuzumab induction.  A total of 7 patients with type 1 diabetes and end-stage renal failure were transplanted with allogenic islets and kidneys procured from brain-dead donors.  To prevent organ rejection, patients received alemtuzumab for induction immunosuppression, followed by sirolimus and tacrolimus.  No glucocorticoids were given at any time.  The median duration of follow-up was 18.3 months (range of 13 to 31).  Kidney survival was 100 %.  Four patients became insulin independent at 1 year; the other 3 reduced insulin use to less than 25 % of the amount required before transplantation.  Serum C-peptide levels were significantly greater post-transplant in all patients, indicating continued islet function.  No major procedure-related complications were observed.  The authors concluded that these findings demonstrated that a steroid-free immunosuppressive regimen consisting of alemtuzumab, sirolimus, and tacrolimus is feasible for simultaneous islet and kidney transplantation.  The question of whether this induction regimen is superior to more standard induction deserves large studies.

Halban et al (2010) stated that beta cell mass and function are decreased to varying degrees in both type 1 and type 2 diabetes.  In the future, islet cell replacement or regeneration therapy may thus offer therapeutic benefit to people with diabetes, but there are major challenges to be overcome.  These researchers performed a review of published peer-reviewed medical literature on beta-cell development and regeneration.  Only publications considered most relevant were selected for citation, with particular attention to the period 2000 to 2009 and the inclusion of earlier landmark studies.  Islet cell regenerative therapy could be achieved by in situ regeneration or implantation of cells previously derived in vitro.  Both approaches are being explored, and their ultimate success will depend on the ability to recapitulate key events in the normal development of the endocrine pancreas to derive fully differentiated islet cells that are functionally normal.  There is also debate as to whether beta-cells alone will assure adequate metabolic control or whether it will be necessary to regenerate islets with their various cell types and unique integrated function.  Any approach must account for the potential dangers of regenerative therapy.  The authors concluded that islet cell regenerative therapy may one day offer an improved treatment of diabetes and potentially a cure.  However, the various approaches are at an early stage of pre-clinical development and should not be offered to patients until shown to be safe as well as more effective than existing therapy.

Cantarelli et al (2013) noted that advances in islet transplantation research have led to remarkable improvements in the outcome in humans with type 1 diabetes.  However, pitfalls, mainly linked both to early liver-specific inflammatory events and to pre-existing and transplant-induced auto- and allo-specific adaptive immune responses, still remain.  In this scenario, research into pancreatic islet transplantation, essential to investigate new strategies to overcome open issues, needs very well-designed pre-clinical studies to obtain consistent and reliable results and select only promising strategies that may be translated into the clinical practice.  These researchers discussed the main shortcomings of the mouse models currently used in islet transplantation research, outlining the main factors and variables to take into account for the design of new pre-clinical studies.  Since several parameters concerning both the graft (i.e., islets) and the recipient (i.e., diabetic mice) may influence transplant outcome, the authors recommended considering several critical points in designing future bench-to-bedside islet transplantation research.

Tekin and colleagues (2016) evaluated short-term and long-term results of the pancreatic islet allotransplantation. Subjects underwent pancreatic islet cell allotransplantation using the Edmonton Protocol; they were followed-up for 10 years after initial islet transplant with up to 3 separate islet infusions. They were given induction treatment using an IL-2R antibody and their maintenance immunosuppression regimen consisted of sirolimus and tacrolimus. A total of 9 patients received a total of 18 islet infusions; 5 patients dropped-out in the early phase of the study. Greater than 50 % drop-out and non-compliance rate resulted from both poor islet function and recurrent side effects of immunosuppression. The remaining 4 (44 %) subjects stayed insulin-free with intervals for at least over 5 years (cumulative time) after the first transplant. Each of them received 3 infusions, on average 445,000 islet equivalent per transplant. Immunosuppression regimen required multiple adjustments in all patients due to recurrent side effects. In the long-term follow-up, kidney function remained stable, and diabetic retinopathy and polyneuropathy did not progress in any of the patients. Patients' panel reactive antibodies remained zero and anti-glutamic acid decarboxylase 65 antibody did not rise after the transplant. Results of metabolic tests including hemoglobin A1c (HbA1c), arginine stimulation, and mixed meal tolerance test were correlated with clinical islet function. The authors concluded that only a small fraction of patients presenting for evaluation were suitable candidates for islet transplantation. Despite thorough patient screening and selection, the drop-out rate was high and was due to combination of poor initial islet graft function and extensive side effects of sirolimus. They stated that immunosuppressant medications must be frequently adjusted to facilitate long-term islet survival and overall health of the islet transplant recipients. Insulin independency was achieved by multiple infusions without detecting PRA. They noted that in properly selected subjects with type 1 DM and severe hypoglycemia with hypoglycemic unawareness, pancreatic islet allotransplantation offered a chance for long-term excellent glycemic control and prevention of progression of diabetic complications, including nephropathy, retinopathy, and neuropathy.

Kaviani et al (2022) noted that islet transplantation has been introduced as a promising approach for the treatment of diabetes mellitus type 1 (DMT1).  Despite many efforts to achieve a perfect treatment, further investigation is still needed to eliminate limitations and challenging issues.  In a systematic review, these researchers examined the effectiveness of islet transplantation in the treatment of patients with DMT1.  They carried out a systematic review on the findings of clinical studies on islet cell transplantation between January 2015 and November 2021.  The search strategy was designed and carried out in PubMed, Cochrane, Scopus, and Web of Science.  Studies have indicated that islet transplantation could reduce the daily insulin requirement as well as improving metabolic stability in diabetic patients.  Even in some patients, insulin independence was achieved during the 1st year of transplantation.  The authors concluded that findings showed that islet transplantation has the potential to become a promising treatment for DMT1.  In this regard, to obtain a trustworthy result, it is essential to design clinical studies (RCTs) with large sample size and long follow-up (cohort studies) to achieve a comprehensive and accurate appraisal of islet transplantation.

In June 2023, the U.S. Food and Drug Administration (FDA) approved Lantidra (donislecel-jujn) (CellTrans, Inc.) allogeneic pancreatic islet cellular suspension for hepatic portal vein infusion for the treatment of adults with type 1 diabetes.

U.S. Food and Drug Administration (FDA)-Approved Indications

  • Lantidra is an allogeneic pancreatic islet cellular therapy indicated for the treatment of adults with type 1 diabetes who are unable to approach target hemoglobin A1C (HbA1c) because of current repeated episodes of severe hypoglycemia despite intensive diabetes management and education. Use in conjunction with concomitant immunosuppression.

    Limitations of Use:

    When considering the risks associated with the infusion procedure and long-term immunosuppression, there is no evidence to show a benefit of administration of Lantidra in patients whose diabetes is well-controlled with insulin therapy or patients with hypoglycemic unawareness who are able to prevent current repeated severe hypoglycemic events (neuroglycopenia requiring active intervention from a third party) using intensive diabetes management (including insulin, devices, and education). Repeated intraportal islet infusions are not recommended in patients who have experienced prior portal thrombosis, unless the thrombosis was limited to second- or third-order portal vein branches. There is no evidence to support the safe and effective use of Lantidra in patients with liver disease, renal failure, or who have received a renal transplant.

Lantidra (donislecel-jujn) is an FDA-approved allogeneic pancreatic islet cellular therapy derived from deceased donor pancreatic cells that are administered as a single infusion into the hepatic portal vein for the treatment of type 1 diabetes. The primary mechanism of action is believed to be the secretion of insulin by the infused (transplanted) allogeneic islet beta cells. For some patients, the infused cells can produce enough insulin, so the patient can be free of daily injections or use of a pump in order to control blood sugar levels. Repeat administration may be warranted depending on the patient’s response to the initial dose. 

FDA approved Lantidra based on the safety and efficacy results from two prospective, open-label, non-randomized, single-arm studies which included a total of 30 adult participants with type 1 diabetes who had hypoglycemic unawareness. Of the 30 participants, 11 subjects received one infusion, 12 received two infusions, and 7 received three infusions of Lantidra. Concomitant immunosuppression study medications were provided (e.g., basiliximab, cyclosporin, mycophenolate mofetil, etanercept, sirolimus, tacrolimus). The outcomes of the combined studies found that, overall, 21 participants did not need to take insulin for a year or more, with 11 participants not needing insulin for one to five years and 10 participants not needing insulin for more than five years. Five participants did not achieve any days of insulin independence, defined as not requiring exogenous insulin to achieve adequate glycemic control. 

Lantidra is contraindicated in patients for whom immunosuppression is contraindicated. Labeled warnings and precautions include risks from concomitant immunosuppression (increased risk of severe infections including opportunistic infections, malignancy, and severe anemia), procedural complications (liver laceration and hemorrhage have occurred), increased risk of graft rejection (patients with a positive T- and B-cell crossmatch between recipient serum and donor lymphocytes may be at increased risk for graft rejection), transmission of donor-derived infections, and panel reactive antibodies (PRA) elevation negatively impacting candidacy for renal transplant. 

Ninety percent (90%) of participants in the clinical studies had at least one serious adverse reaction. The major causes were attributed to:

  • Infusion procedure

    • liver laceration/hematoma, hemorrhage, and intra-abdominal bleeding (13%)
    • elevation of portal pressure (7%)
  • Immunosuppression

    • Infection (87%)
    • Malignancy (37%)

Some adverse reactions to the therapy required participants to discontinue immunosuppressive medicines, which ended their insulin independence. Concomitant use of immunosuppression is required to maintain islet cell viability, preventing the body from destroying the infused (transplanted) cells.

Pancreatic Islet Xenotransplantation

Xenotransplantation involves implantation or infusion into a human receipient of "either (a) live cells, tissues, or organs from a nonhuman animal source, or (b) human body fluids, cells, tissues or organs that have had ex vivo contact with live nonhuman animal cells, tissues or organs" (FDA, 2021).

Marigliano et al (2011) stated that the therapy of type 1 diabetes is an open challenging problem.  The restoration of normoglycemia and insulin independence in immunosuppressed type 1 diabetic recipients of islet allotransplantation has shown the potential of a cell-based diabetes therapy.  Even if successful, this approach poses a problem of scarce tissue supply.  Xenotransplantation can be the answer to this limited donor availability and, among possible candidate tissues for xenotransplantation, porcine islets are the closest to a future clinical application.  Xenotransplantation, with pigs as donors, offers the possibility of using healthy, living, and genetically modified islets from pathogen-free animals available in unlimited number of islets.  Several studies in the pig to non-human primate model demonstrated the feasibility of successful pre-clinical islet xenotransplantation and have provided insights into the critical events and possible mechanisms of immune recognition and rejection of xenogeneic islet grafts.  Particularly promising results in the achievement of prolonged insulin independence were obtained with newly developed, genetically modified pigs islets able to produce immunoregulatory products, using different implantation sites, and new immunotherapeutic strategies.  Nonetheless, the authors concluded that further efforts are needed to generate additional safety and efficacy data in non-human primate models to safely translate these findings into the clinic.

Samy et al (2014) stated that type I diabetes remains a significant clinical problem in need of a reliable, generally applicable solution.  Both whole organ pancreas and islet allo-transplantation have been shown to grant patients insulin independence, but organ availability has restricted these procedures to an exceptionally small subset of the diabetic population.  Porcine islet xenotransplantation has been pursued as a potential means of overcoming the limits of allo-transplantation, and several pre-clinical studies have achieved near-physiologic function and year-long survival in clinically relevant pig-to-primate model systems.  These proof-of-concept studies have suggested that xenogeneic islets may be poised for use in clinical trials. 

Matsumoto and associates (2016) noted that while islet allotransplantation has become a viable option for the treatment of unstable type 1 diabetes (T1DM), donor shortage and the necessity of the immunosuppressive drugs are 2 main obstacles.  To solve these issues, these researchers performed islet xenotransplantation using encapsulated neonatal porcine islets without immunosuppressive drugs.  Two different doses (approximately 5,000 IEQ/kg and 10,000 IEQ/kg) of encapsulated neonatal porcine islets were transplanted twice (total approximately 10,000 IEQ/kg and 20,000 IEQ/kg) into 4 T1DM patients in each group (total 8 patients).  In the higher dose group, all 4 patients improved HbA1c.  This was maintained at a level of less than 7 % for greater than 600 days with significant reduction of the frequency of unaware hypoglycemic events.  These investigators did not evaluate the maturity of neonatal islets before transplantation, however; they plan to evaluate the maturity of neonatal islets before transplantation for the next clinical trial.  The authors concluded that encapsulated neonatal porcine islet xenotransplantation could maintain HbA1c less than 7 % with significant reduced hypoglycemic events without immunosuppression greater than 600 days.  They believed this study is the prologue for the clinical xenotransplantation to solve the issue of donor shortage.

Pancreas Transplantation With Exocrine Drainage Through a Duodenoduodenostomy Versus Duodenojejunostomy

Lindahl and colleagues (2018) noted that until recently, pancreas transplantation has mostly been performed with exocrine drainage via duodenojejunostomy (DJ).  Since 2012, DJ was substituted with duodenoduodenostomy (DD) in the authors’ hospital, allowing endoscopic access for biopsies.  This study assessed safety profiles with DD versus DJ procedures and clinical outcomes with the DD technique in pancreas transplantation.  DD patients (n = 117; 62 simultaneous pancreas-kidney [SPKDD ] and 55 pancreas transplantation alone [PTADD ] with median follow-up of 2.2 years) were compared with DJ patients (n = 179; 167 SPKDJ and 12 PTADJ ) transplanted in the period 1998 to 2012 (pre-DD era).  Post-operative bleeding and pancreas graft vein thrombosis requiring re-laparotomy occurred in 17 % and 9 % of DD patients versus 10 % (p = 0.077) and 6 % (p = 0.21) in DJ patients, respectively.  Pancreas graft rejection rates were still higher in PTADD patients versus SPKDD patients (p = 0.003).  Hazard ratio (HR) for graft loss was 2.25 (95 % confidence interval [CI]: 1.00 to  5.05; p = 0.049) in PTADD versus SPKDD recipients.  The authors concluded that compared with the DJ procedure, the DD procedure did not reduce post-operative surgical complications requiring re-laparatomy or improve clinical outcomes after pancreas transplantation despite serial pancreatic biopsies for rejection surveillance.  It remains to be seen whether better rejection monitoring in DD patients translates into improved long-term pancreas graft survival.

Pantoprazole plus Sitagliptin for Restoration of Insulin Independence in Islet Transplant Recipients with Early Graft Insufficiency

Senior and associates (2017) stated that resuming insulin use due to waning function is common after islet transplantation.  Animal studies suggested that gastro-intestinal (GI) hormones, including gastrin and incretins may increase β-cell mass.  In a single-center, uncontrolled, open-label, pilot study, these researchers tested the hypothesis that pantoprazole plus sitagliptin (pantoprazole 40 mg twice- plus daily sitagliptin 100 mg daily for 6 months), would restore insulin independence in islet transplant recipients with early graft insufficiency and determined whether this would persist after a 3-month washout.  After 6 months of treatment, 2 of 8 participants (25 %) achieved the primary end-point, defined as HbA1c of less than 42 mmol/mol (6 %), fasting plasma glucose of less than 7.0 mmol, C-peptide of greater than 0.5 nmol and no insulin use.  There was a significant reduction in mean insulin dose, but no change in HbA1C or weight.  There were no changes in the acute insulin response to arginine, the mixed meal tolerance test or blinded continuous glucose monitoring.  After the washout, no participants met the primary end-point and HbA1C increased from 45 ± 8 mmol/mol (6.3 ± 0.7 %) to 51 ± 6 mmol/mol (6.8 ± 0.6 %) (p < 0.05); 2 participants had mild-moderate transient GI side effects.  There were no episodes of hypoglycemia.  The authors concluded that sitagliptin plus pantoprazole was well-tolerated and safe and may restore insulin independence in some islet transplant recipients with early graft insufficiency, but this was not sustained when treatment was withdrawn.  They stated that a larger, controlled trial is needed to confirm the effectiveness of this combination to achieve insulin independence and to confidently exclude any persistent benefit for graft function.

Total Pancreatectomy with Islet Autotransplantation for the Treatment of Chronic Pancreatitis

Berman and co-workers (2019) noted that painful chronic pancreatitis (CP) is the main indication for analgesic pancreatectomy with simultaneous islet autotransplantation to prevent post-operative diabetes mellitus (DM). However, advanced CP may lead to insulin secretion disorders and DM. There are doubts as to whether islet autotransplantation in such cases is an appropriate procedure. These researchers analyzed the findings of islet autotransplantation in patients with CP with already diagnosed with DM.  Between 2008 and 2015, at the Department of General and Transplantation Surgery, patients with CP and unsatisfying pain treatment with positive fasting C-peptide (greater than 0.3 ng/ml) level were qualified for simultaneous pancreatectomy and islet autotransplantation; 8 procedures were performed.  In 5 cases, patients had DM diagnosed prior to the procedure (DM group n = 5); 3 patients without DM diagnosed prior to surgery were the control group (n = 3).  There were no cases of procedure-related deaths in either group. Pain relief without analgesics was reported by all patients. Good islet function was observed in 80 % (4/5) of the DM group versus 100 % (3/3) in the control group (p = ns). Brittle diabetes was diagnosed in 1 patient in the DM group as a result of islet primary non-function.  The authors concluded that patients with CP-related severe pain and DM patients with positive C-peptides should be considered for pancreatectomy and islet autotransplantation.

Kempeneers and associates (2019) stated that the rationale for total pancreatectomy in painful, treatment refractory CP is pain control. Concomitant islet cell autotransplantation could prevent the loss of islet cell function. In a systematic review and meta-analysis, these researchers examined the impact of total pancreatectomy with islet autotransplantation (TPIAT) on pain and quality of life (QOL). This meta-analysis was carried out according the Meta-analyses of Observational Studies in Epidemiology guideline. The Cochrane Library, PubMed, and Embase were searched for the following terms (1990 through April 2018): total pancreatectomy and chronic pancreatitis. Studies were included when addressing TPIAT for chronic pancreatitis in adults. Studies that reported no data on pain, endocrine function, or QOL were excluded. Quality was assessed using the Newcastle-Ottawa scale for evaluation of all studies. These investigators included 15 observational studies evaluating 1,255 patients, of whom 28 % had had endoscopic and 23 % operative therapy. One year after TPIAT, the opioid-free rate had improved from between 0 % and 15 % to 63 % (95 % CI: 46 to 77), and the insulin-free rate had decreased from between 89.5 % and 100 % to 30 % (95 % CI: 20 to 43). An alcoholic etiology was associated with a lesser insulin-free rate after TPIAT; QOL improved statistically after TPIAT.  Publication bias was present for both opioid and insulin outcomes.  The authors concluded that in selected patients with painful, treatment refractory CP, evidence showed that TPIAT was effective for pain control in almost 2/3 of patients, whereas the insulin-free rate was relatively low.

Abu-El-Haija and colleagues (2020) noted that advances in the understanding of TPIAT have been made.  These investigators defined indications and outcomes of TPIAT. Expert physician-scientists from North America, Asia, and Europe reviewed the literature to address 6 questions selected by the writing group as high priority topics.  A consensus was reached by voting on statements generated from the review.  Consensus statements were voted upon with strong agreement reached that (Q1) TPIAT may improve QOL, reduce pain and opioid use, and potentially reduce medical utilization; that (Q3) TPIAT offers glycemic benefit over TP alone; that (Q4) the main indication for TPIAT is disabling pain, in the absence of certain medical and psychological contraindications; and that (Q6) islet mass transplanted and other disease features may impact DM outcomes. Conditional agreement was reached that (Q2) the role of TPIAT for all forms of CP is not yet identified and that head-to-head comparative studies are lacking, and that (Q5) early surgery is likely to improve outcomes as compared to late surgery.  The authors concluded that agreement on TPIAT indications and outcomes has been reached through this working group; further studies are needed to answer the long-term outcomes and maximize efforts to optimize patient selection.

Ex-Situ Machine Pancreas Perfusion (in Hypothermic or Normothermic Settings) for Evaluation of Pancreas Before Transplantation

Prudhomme and colleagues (2020) noted that pancreas transplantation is currently one of the best treatments proposed in highly selected patients with unstable and brittle T1DM.  The goal of pancreas transplantation is to restore normoglycemia and avoid the occurrence of complications associated with diabetes.  Graft pancreatitis and thrombosis, arising from ischemia reperfusion injuries, are major causes of graft loss in the post-operative period.  Ex-situ perfusion, in hypothermic or normothermic settings, allowed to improve ischemic reperfusion injury in other organ transplantations (e.g., kidney, liver, or lung).  The development of pancreatic graft perfusion techniques would limit these ischemic re-perfusion injuries.  In a systematic review, these researchers examined the safety and feasibility of ex-situ perfusion of pancreas for whole-organ transplantation.  English literature regarding pancreas perfusion was analyzed using electronic database Medline via PubMed (1950 to 2018).  Exclusion criteria were studies that did not specify the technical aspects of machine perfusion and studies focused only on pancreas perfusion for islet isolation.  Hypothermic machine perfusion for pancreas preservation has been evaluated in 9 studies and normothermic machine perfusion in 10 studies.  These investigators examined machine perfusion model, types of experimental model, anatomy, perfusion parameters, flushing and perfusion solution, length of perfusion, and comparison between static cold storage (SCS) and perfusion.  The authors concluded that this review compared ex-situ machine perfusion of experimental pancreas for whole-organ transplantation.  Pancreas perfusion was feasible and could be a helpful tool to examine pancreas before transplantation.  These researchers stated that pancreas perfusion (in hypothermic or normothermic settings) could reduce ischemic re-perfusion injuries, and maybe could avoid pancreas thrombosis and reduce morbidity of pancreas transplantation.

These researchers stated that further experimentation in animal models is needed to confirm the protective effect of pancreas perfusion, to select precise perfusion parameters useful for pancreas evaluation, and to explain the mechanisms of tissue protection.  These investigators could then consider the use of pancreas perfusion for human pancreas to broaden the organ pool and improve outcomes.  In the future, it should be important to examine ischemia-reperfusion injuries by model of transplantation and to invest in NP technology as has been performed in liver and kidney transplantation.  Indeed, with future development of portable NP machine devices, cold ischemia time can be minimized as the organ does not need to travel.  Moreover, advantage of NP machine is the possibility to treat the organ (accommodation) prior to transplantation, instead of treating the recipient.

The First World Consensus Conference on Pancreas Transplantation (Boggi et al, 2021a) concluded: "No recommendation was drawn on the use of machine perfusion because of lack of clinical studies."

IGL-1 Preservation Solution for Pancreas Transplantation

In a systematic review, Habran and colleagues (2020) evaluated published data on the effectiveness of Institut Georges Lopez-1 (IGL-1) as a preservation solution for kidney and pancreas grafts.  These researchers carried out a literature search of PubMed, Embase, Web of Science, and the Cochrane Library databases.  Human studies examining the effects of IGL-1 preservation solution in kidney and/or pancreas transplantation were included.  Outcome data on kidney and pancreas graft function were extracted.  Of 1,513 unique articles identified via the search strategy, 4 articles could be included in the systematic review.  Of these, 2 retrospective studies reported on the outcome of IGL-1 compared to University of Wisconsin (UW) solution in kidney transplantation.  These studies showed kidneys preserved in IGL-1 had improved early function (2 weeks post-transplant) compared to UW.  Follow-up was limited to 1 year and showed similar graft and patient survival rates when reported.  Two case-series studies described acceptable early outcomes (up to 1 month) of simultaneous kidney / pancreas transplantation after storage in IGL-1.  As only 4 clinical studies were identified, these investigators widened their search to include 4 eligible large animal studies; 3 compared IGL-1 with UW in ovine renal transplant models with inconclusive or mildly positive results.  One ovine pancreas transplant study suggested better early outcome with IGL-1 compared to UW.  Too few published data were available to allow any firm conclusions to be drawn regarding the effectiveness of IGL-1 as a preservation solution of kidney and pancreas grafts.  The authors concluded that the limited published clinical data appeared to suggest that IGL-1 was a safe and promising preservation solution for the static cold storage of kidney and pancreas grafts; however, because there are no large data series available.  It is unclear whether outcomes after transplantation of kidneys and pancreas preserved with IGL-1 are equivalent to those obtained with UW or other commonly used abdominal preservation solutions.  Moreover, these researchers stated that additional well-designed studies and, ideally randomized controlled trials (RCTs), are needed to demonstrate equivalence or superiority of one solution over the other.

The First World Consensus Conference on Pancreas Transplantation (Boggi et al, 2021b) stated that "no conclusion could be drawn on Institut Georges Lopez-1 (IGL-1) solution, because of lack of a comparison group in available studies."


Glossary of Terms

Table: Glossary of Terms
Term Definition
Allogeneic Cells or tissues obtained from a donor of the same species
Allotransplantation Transplantation of organs, tissues, cells from a donor
Autologous Cells or tissues obtained from same individual
Autotransplantation Transplantation of organs, tissues, cells from one part of the body to another in the same individual
Labile, brittle Liable to change, unstable
Xenotransplantation Non-human cells, tissues, or organs used to treat medical conditions in humans

References

The above policy is based on the following references:

  1. Abu-El-Haija M, Anazawa T, Beilman GJ, et al. The role of total pancreatectomy with islet autotransplantation in the treatment of chronic pancreatitis: A report from the International Consensus Guidelines in chronic pancreatitis. Pancreatology. 2020;20(4):762-771.
  2. Amara D, Hansen KS, Kupiec-Weglinski SA, et al. Pancreas transplantation for type 2 diabetes: A systematic review, critical gaps in the literature, and a path forward. Transplantation. 2022;106(10):1916-1934.
  3. Atkinson MA, Eisenbarth GS. Type 1 diabetes: New perspectives on disease pathogenesis and treatment. Lancet. 2001;358(9277):221-229.
  4. Balamurugan AN, Bottino R, Giannoukakis N, Smetanka C. Prospective and challenges of islet transplantation for the therapy of autoimmune diabetes. Pancreas. 2006;32(3):231-243.
  5. Barr ML, Belghiti J, Villamil FG, et al. A report of the Vancouver Forum on the care of the live organ donor: Lung, liver, pancreas, and intestine data and medical guidelines. Transplantation. 2006;81(10):1373-1385.
  6. Benedetti E, Troppmann C, Gruessner AC, et al. Pancreas graft loss caused by intra-abdominal infection. A risk factor for a subsequent pancreas retransplantation. Arch Surg. 1996;131(10):1054-1060.
  7. Berman A, Pawelec K, Fiedor P. Allogeneic transplantation of isolated islet cells in clinical practice. Pol Arch Med Wewn. 2009;119(5):326-332.
  8. Berman A, Wszola M, Gorski L, et al. Islet autotransplantation in diabetic patients. Transplant Proc. 2019;51(8):2781-2786.
  9. Bertuzzi F, Ricordi C. Prediction of clinical outcome in islet allotransplantation. Diabetes Care. 2007;30(2):410-417.
  10. BlueCross BlueShield Association (BCBSA), Technology Evaluation Center (TEC). Pancreas retransplantation. TEC Assessment Program. Chicago IL: BCBSA; April 2002;16(23).
  11. Boggi U, Vistoli F, Andres A, et al. First World Consensus Conference on pancreas transplantation: Part II - recommendations. Am J Transplant. 2021a;21 Suppl 3(Suppl 3):17-59.
  12. Boggi U, Vistoli F, Marchetti P, et al.; World Consensus Group on Pancreas Transplantation. First world consensus conference on pancreas
    transplantation: Part I - Methods and results of literature search. Am J Transplant. 2021b;21 Suppl 3(Suppl 3):1-16.
  13. Bohman SO, Tyden G, Wilezek A, et al. Prevention of kidney graft diabetic nephropathy by pancreas transplantation in man. Diabetes. 1985;34:306-308.
  14. Bohman SO, Wilczek H, Jaremko G, et al. Recurrence of diabetic nephropathy in human renal allografts: Preliminary report of a biopsy study. Transplant Proc. 1984;16:649-653.
  15. Boudreau R, Hodgson A. Pancreas transplantation to restore glucose control: Review of clinical and economic evidence. Technology Report No. 84. Ottawa, ON: Canadian Agency for Drugs and Technologies in Health (CADTH); 2007.
  16. Bramis K, Gordon-Weeks AN, Friend PJ, et al. Systematic review of total pancreatectomy and islet autotransplantation for chronic pancreatitis. Br J Surg. 2012;99(6):761-766.
  17. Bretzel RG, Brendel M, Eckhard M, et al. Islet transplantation: present clinical situation and future aspects. Exp Clin Endocrinol Diabetes. 2001;109 Suppl 2:S384-S399.
  18. Burke GW, Ciancio G, Sollinger HW. Advances in pancreas transplantation. Transplantation. 2004;77(9 Suppl):S62-S67.
  19. Cantarelli E, Citro A, Marzorati S, et al. Murine animal models for preclinical islet transplantation: No model fits all (research purposes). Islets. 2013;5(2):79-86.
  20. CellTrans Inc. Lantidra (donislecel-jujn) allogeneic pancreatic islet cellular suspension for hepatic portal vein infusion. Prescribing Information. Chicago, IL: CellTrans; June 2023.
  21. Centers for Medicare and Medicaid Services (CMS). Decision memo for pancreas transplants (CAG-00295R). Medicare Coverage Database. Baltimore, MD: CMS; April 26, 2006.
  22. Clayton HA, Davies JE, Pollard CA, et al. Pancreatectomy with islet autotransplantation for the treatment of severe chronic pancreatitis: The first 40 patients at the Leicester General Hospital. Transplantation. 2003;76(1):92-98.
  23. Cooper DK, Bottino R. Recent advances in understanding xenotransplantation: Implications for the clinic. Expert Rev Clin Immunol. 2015;11(12):1379-1390.
  24. Cooper DK, Casu A. The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes -- chapter 4: Pre-clinical efficacy and complication data required to justify a clinical trial. Xenotransplantation. 2009;16(4):229-238.
  25. Dong M, Parsaik AK, Erwin PJ, et al. Systematic review and meta-analysis: Islet autotransplantation after pancreatectomy for minimizing diabetes. Clin Endocrinol. 2011;75(6):771-779.
  26. Dubernard JM, Tajra LC, Lefrancois N, et al. Pancreas transplantation: Results and indications. Diabetes Metab. 1998;24:195-199.
  27. Eriksson O, Eich T, Sundin A, et al. Positron emission tomography in clinical islet transplantation. Am J Transplant. 2009;9(12):2816-2824.
  28. Federlin KF, Jahr H, Bretzel RG. Islet transplantation as treatment of type 1 diabetes: From experimental beginnings to clinical application. Exp Clin Endocrinol Diabetes. 2001;109 Suppl 2:S373-S383.
  29. Froud T, Faradji RN, Pileggi A, et al. The use of exenatide in islet transplant recipients with chronic allograft dysfunction: Safety, efficacy, and metabolic effects. Transplantation. 2008;86(1):36-45.
  30. Gangemi A, Salehi P, Hatipoglu B, et al. Islet transplantation for brittle type 1 diabetes: The UIC protocol. Am J Transplant. 2008;8(6):1250-1261.
  31. Genzini T, Crescentini F, Torricelli FC, et al. Pancreas retransplantation: Outcomes of 20 cases. Transplant Proc. 2006;38(6):1937-1938.
  32. Gruessner AC, Sutherland DE. Pancreas transplant outcomes for United States (US) cases reported to the United Network for Organ Sharing (UNOS) and non-US cases reported to the International Pancreas Transplant Registry (IPTR) as of October 2000. Clin Transpl. 2000;45-72.
  33. Guo B, Corabian P, Harstall C. Islet transplantation for the treatment of type 1 diabetes: An update. IHE Report. Edmonton, AB: Institute of Health Economics (IHE); December 2008.
  34. Guo B, Harstall C, Corabian P. Islet cell transplantation for the treatment of non-uremic type 1 diabetic patients with severe hypoglycemia. Health Technology Assessment. HTA 31. Edmonton, AB: Alberta Heritage Foundation for Medical Research (AHFMR); April 2003.
  35. Habran M, De Beule J, Jochmans I. IGL-1 preservation solution in kidney and pancreas transplantation: A systematic review. PLoS One. 2020;15(4):e0231019.
  36. Halban PA, German MS, Kahn SE, Weir GC. Current status of islet cell replacement and regeneration therapy. J Clin Endocrinol Metab. 2010;95(3):1034-1043.
  37. Hering BJ, Cozzi E, Spizzo T, et al. First update of the International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes -- Executive summary. Xenotransplantation. 2016;23(1):3-13.
  38. Hering BJ, Kandaswamy R, Ansite JD, et al. Single-donor, marginal-dose islet transplantation in patients with type 1 diabetes. JAMA. 2005;293(7):830-835.
  39. Hering BJ, O'Connell PJ. First update of the International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes -- Chapter 6: Patient selection for pilot clinical trials of islet xenotransplantation. Xenotransplantation. 2016;23(1):60-76.
  40. Hermann M, Margreiter R, Hengster P. Human islet autotransplantation: The trail thus far and the highway ahead. Adv Exp Med Biol. 2010;654:711-724.
  41. Humar A, Kandaswamy R, Drangstveit MB, et al. Surgical risks and outcome of pancreas retransplants. Surgery. 2000;127(6):634-640.
  42. Institute for Clinical Systems Improvement (ICSI). Pancreas transplant for insulin-dependent diabetes. ICSI Technology Assessment Report No. 4. Bloomington, MN: ICSI; updated October 2003.
  43. Jie T, Hering BJ, Ansite JD, et al. Pancreatectomy and auto-islet transplant in patients with chronic pancreatitis [abstract]. Surgical Forum, Alimentary Tract Category. #2005-466. American College of Surgeons 91st Annual Clinical Congress, San Francisco, CA, October 16-20, 2005.
  44. Katz H, Homan M, Velosa J, et al. Effects of pancreas transplantation on postprandial glucose metabolism. N Engl J Med. 1991;325:1278-1283.
  45. Kaviani M, Geramizadeh B, Karimzadeh S, et al. Effectiveness of islet transplantation in diabetes mellitus type 1: A systematic review of the recent evidences. Minerva Endocrinol (Torino). 2022 Sep 30 [Online ahead of print].
  46. Kempeneers MA, Scholten L, Verkade CR, et al; Dutch Pancreatitis Study Group. Efficacy of total pancreatectomy with islet autotransplantation on opioid and insulin requirement in painful chronic pancreatitis: A systematic review and meta-analysis. Surgery. 2019;166(3):263-270.
  47. Kennedy WR, Navarro X, Goetz FC, et al. Effects of pancreatic transplantation on diabetic neuropathy. N Engl J Med. 1990;322:1031-1037.
  48. Kiberd BA, Larson T. Estimating the benefits of solitary pancreas transplantation in nonuremic patients with type 1 diabetes mellitus: A theoretical analysis. Transplantation. 2000;70(7):1121-1127.
  49. Korsgren O, Nilsson B, Berne C, et al. Current status of clinical islet transplantation. Transplantation. 2005;79(10):1289-1293.
  50. Kuo PC, Johnson LB, Schweitzer EJ, et al. Solitary pancreas allografts. The role of percutaneous biopsy and standardized histologic grading of rejection. Arch Surg. 1997;132(1):52-57.
  51. Lindahl JP, Horneland R, Nordheim E, et al. Outcomes in pancreas transplantation with exocrine drainage through a duodenoduodenostomy versus duodenojejunostomy. Am J Transplant. 2018;18(1):154-162.
  52. Marigliano M, Bertera S, Grupillo M, et al. Pig-to-nonhuman primates pancreatic islet xenotransplantation: An overview. Curr Diab Rep. 2011;11(5):402-412.
  53. Matsumoto S, Abalovich A, Wechsler C, et al. Clinical benefit of islet xenotransplantation for the treatment of type 1 diabetes. EBioMedicine. 2016;12:255-262.
  54. Mineo D, Ricordi C, Xu X, et al. Combined islet and hematopoietic stem cell allotransplantation: A clinical pilot trial to induce chimerism and graft tolerance. Am J Transplant. 2008;8(6):1262-1274.
  55. Najarian JS, Kaufman DB, Fryd DS, et al. Long-term survival following kidney transplantation in 100 type 1 diabetic patients. Transplantation. 1989;1:106-113.
  56. National Institute for Clinical Excellence (NICE). Pancreatic islet cell transplantation. Interventional Procedure Guidance 13. London, UK: NICE; October 2003.
  57. O'Connell PJ. The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes -- chapter 6: Patient selection for pilot clinical trials of islet xenotransplantation. Xenotransplantation. 2009;16(4):249-254.
  58. Odorico JS, Becker YT, Groshek M, et al. Improved solitary pancreas transplant graft survival in the modern immunosuppressive era. Cell Transplant. 2000;9(6):919-927.
  59. Odorico JS, Leverson GE, Becker YT, et al. Pancreas transplantation at the University of Wisconsin. Clin Transpl. 1999;199-210.
  60. Ollinger R, Margreiter C, Bosmuller C, et al. Evolution of pancreas transplantation: Long-term results and perspectives from a high-volume center. Ann Surg. 2012;256(5):780-786; discussion 786-787.
  61. Ontario Ministry of Health and Long-Term Care, Medical Advisory Secretariat. Islet transplantation. Health Technology Scientific Literature Review. Toronto, ON: Ontario Ministry of Health and Long-Term Care; October 2003.
  62. Pichon Riviere A, Augustovski F, Alcaraz A, et al. Pancreas islet stem cells and transplant for the treatment of diabetes mellitus [summary].  IRR No. 93. Buenos Aires, Argentina: Institute for Clinical Effectiveness and Health Policy (IECS); 2006.
  63. Piper M, Seidenfeld J, Aronson N. Islet transplantation in patients with type 1 diabetes mellitus. Evidence Report/Technology Assessment No. 98. (Prepared by Blue Cross and Blue Shield Association Technology Evaluation Center Evidence-based Practice Center under Contract No. 290-02-0026). AHRQ Publication 04-E017-2. Rockville, MD: Agency for Healthcare Research and Quality (AHRQ); August 2004.
  64. Prudhomme T, Kervella D, Le Bas-Bernardet S, et al. Ex situ perfusion of pancreas for whole-organ transplantation: Is it safe and feasible? A systematic review. J Diabetes Sci Technol. 2020;14(1):120-134.
  65. Ramsay RC, Goetz FC, Sutherland DER, et al. Progression of diabetic retinopathy after pancreas transplantation for insulin-dependent diabetes mellitus. N Engl J Med. 1988;318:208-214.
  66. Reddy KS, Shokouh-Amiri H, Stratta RJ, et al. Successful reuse of portal-enteric technique in pancreas retransplantation. Transplantation. 2000;69(11):2443-2445.
  67. Robertson RP. Pancreatic islet transplantation for diabetes: Successes, limitations, and challenges for the future. Mol Genet Metab. 2001;74(1-2):200-205.
  68. Robertson RP, Davis C, Larsen J, et al. Pancreas and islet transplantation for patients with diabetes. Diabetes Care. 2003;26 Suppl:S120.
  69. Robertson RP, Davis C, Larsen J, et al. Pancreas transplantation in type 1 diabetes. Diabetes Care. 2004;27(Suppl 1):S105.
  70. Rodriguez Rilo HL, Ahmad SA, D'Alessio D, et al. Total pancreatectomy and autologous islet cell transplantation as a means to treat severe chronic pancreatitis. J Gastrointest Surg. 2003;7;978-989.
  71. Ruggles JA, Kelemen D, Baron A. Emerging therapies: Controlling glucose homeostasis, immunotherapy, islet transplantation, gene therapy, and islet cell neogenesis and regeneration. Endocrinol Metab Clin North Am. 2004;33(1):239-252, xii.
  72. Ryan EA, Bigam D, Shapiro AM. Current indications for pancreas or islet transplant. Diabetes Obes Metab. 2006;8(1):1-7.
  73. Samy KP, Martin BM, Turgeon NA, Kirk AD. Islet cell xenotransplantation: A serious look toward the clinic. Xenotransplantation. 2014;21(3):221-229.
  74. Sansalone CV, Maione G, Rossetti O, et al. Pancreas retransplantation: Ideal timing and early and late results. Transplant Proc. 2006;38(4):1153-1155.  
  75. Sasaki TM, Gray RS, Ratner RE, et al. Successful long-term kidney-pancreas transplants in diabetic patients with high C-peptide levels. Transplantation. 1998;65:1510-1512.
  76. Saudek F, Girman P, Kríz J, et al. Islet transplantation for treatment of type-1 diabetes mellitus. Cas Lek Cesk. 2011;150(1):49-55.
  77. Scalea JR, Pettinato L, Fiscella B, et al. Successful pancreas transplantation alone is associated with excellent self-identified health score and glucose control: A retrospective study from a high-volume center in the United States. Clin Transplant. 2018;32(2).
  78. Senior PA, Koh A, Yau J, et al. Sitagliptin plus pantoprazole can restore but not maintain insulin independence after clinical islet transplantation: Results of a pilot study. Diabet Med. 2017;34(2):204-212.
  79. Shapiro AM, Lakey JR. Future trends in islet cell transplantation. Diabetes Technol Ther. 2000;2(3):449-452.
  80. Shapiro AM, Ryan EA, Lakey JR. Pancreatic islet transplantation in the treatment of diabetes mellitus. Best Pract Res Clin Endocrinol Metab. 2001;15(2):241-264.
  81. Sollinger HW, Odorico JS, Knechtle SJ, et al. Experience with 500 simultaneous pancreas-kidney transplants. Ann Surg. 1998;228:284-296.
  82. Stock PG, Bluestone JA. Beta-cell replacement for type I diabetes. Annu Rev Med. 2004;55:133-156.
  83. Stratta RJ. Immunosuppression in pancreas transplantation: Progress, problems and perspective. Transpl Immunol. 1998;6(2):69-77.
  84. Stratta RJ. Review of immunosuppressive usage in pancreas transplantation. Clin Transplant. 1999;13(1 Pt 1):1-12.
  85. Stratta RJ, Gaber AO, Shokouh-Amiri MH, et al. A prospective comparison of systemic-bladder versus portal-enteric drainage in vascularized pancreas transplantation. Surgery. 2000;127(2):217-226.
  86. Stratta RJ, Lowell JA, Sudan D, et al. Retransplantation in the diabetic patient with a pancreas allograft. Am J Surg. 1997;174(6):759-763.
  87. Sutherland DE, Gruessner RW, Dunn DL, et al. Lessons learned from more than 1,000 pancreas transplants at a single institution. Ann Surg. 2001;233(4):463-501.
  88. Tan J, Yang S, Cai J, et al. Simultaneous islet and kidney transplantation in seven patients with type 1 diabetes and end-stage renal disease using a glucocorticoid-free immunosuppressive regimen with alemtuzumab induction. Diabetes. 2008;57(10):2666-2671.
  89. Tekin Z, Garfinkel MR, Chon WJ, et al. Outcomes of pancreatic islet allotransplantation using the Edmonton Protocol at the University of Chicago. Transplant Direct. 2016;2(10):e105.
  90. U.S. Food and Drug Administration (FDA). FDA approves first cellular therapy to treat patients with type 1 diabetes. FDA News Release. Silver Spring, MD: FDA; June 29, 2023.
  91. U.S. Food and Drug Administration (FDA). Xenotransplantation [website]. Silver Spring, MD: FDA; March 3, 2021. Available at: https://www.fda.gov/vaccines-blood-biologics/xenotransplantation. Accessed July 31, 2023.
  92. Van der Vliet JA, Navarro X, Kennedy WR, et al. The effect of pancreas transplantation on diabetic polyneuropathy. Transplantation. 1988;45:368-370.
  93. Vardanyan M, Parkin E, Gruessner C, Rodriguez Rilo HL. Pancreas vs. islet transplantation: A call on the future. Curr Opin Organ Transplant. 2010;15(1):124-130.
  94. Weegman BP, Taylor MJ, Baicu SC, et al. Plasticity and aggregation of juvenile porcine islets in modified culture: Preliminary observations. Cell Transplant. 2016;25(10):1763-1775.
  95. White SA, Kimber R, Veitch PS, et al. Surgical treatment of diabetes mellitus by islet cell and pancreas transplantation. Postgrad Med J. 2001;77(908):383-387.