throbber
CLEVELAND CLIN!’C FOUNDA:T~ON
`
`COOPER CLINIC
`
`GEISINGER MEDICAL CEN’FE,R
`
`GUTHRIECLINIC
`
`HENRY FORD HOSPITAl:.
`
`JO’SLIN,CLINtC
`
`LAHEY CLINIC FOUNDATION
`
`LOVELACE MEDICAL CEN’,TER
`
`MARSNFIELD CLINIC
`
`MAYO;,CLINtC. ,=
`
`OCHSNER CLINIC
`
`PALO ALTO MEDICAL CLINIC
`
`SCOTT AND WHITE CLINIC
`~CRIPPS ~LINIC AND RESEARCH FOUNDATION
`
`Sandoz Inc.
`Exhibit 1019-0001
`
`JOINT 1019-0001
`
`

`
`urrent
`lin
`Pract ic e
`
`FRANZ H. MESSERLI, M.D.
`EDITOR
`
`1987
`W.B. SAUNDERS COMPANY
`Philadelphia London Toronto
`Sydney Tokyo Hong Kong
`
`Sandoz Inc.
`Exhibit 1019-0002
`
`JOINT 1019-0002
`
`

`
`W. B. Saunders Company: West Washington Square
`Philadelphia, PA 19105
`
`Library of Congress Cataloging in Publication Data
`
`Current clinical practice
`
`1. Internal medicine. 2. Medicine, Clinical.
`L Messerli, Franz H. [DNLM: 1. Medicine. WB 100 C976]
`
`RC46. C95 1987 616 86-6512
`
`ISBN 0-7216-1460-4
`
`Editor: John Dyson
`
`Developmental Editor: Carole Morrison
`
`Designer: Terri Siegel
`
`Production Manager: Bill Preston
`
`Manuscript Editor: David Harvey
`
`Illustration Coordinator: Walt Verbitski
`
`Indexer: Ella Shapiro
`
`Current Clinical Practice
`
`ISBN 0-7216-1460-4
`
`© 1987 by W. B. Saunders Company. Copyright under the Uniform Copyright Convention. Simul-
`taneously published in Canada. All rights reserved. This book is. protected by copyright: No part of it
`may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, eJectronic,
`mechanical, photocopying, recording, or otherwise, without written permission from the publisher. Made
`in the United States of America. Library of Congress catalog card number 86-651Z
`
`Last digit is the print number: 9 8 7 6 5 4 3 2 1
`
`Sandoz Inc.
`Exhibit 1019-0003
`
`JOINT 1019-0003
`
`

`
`BLOOD AND
`NEOPLASTIC DISORDERS
`
`ERNEST BEUTLER
`JAMES K. WEICK
`
`1 . BONE MARROW
`TRANSPLANTATION IN
`
`ANEMIA
`Wayne Spruce
`SCRIPPS CLINIC AND RESEARCH FOUNDATION
`
`ETIOLOGIC CONSIDERATIONS
`
`Until the advent of bone marrow transplantation,
`severe aplastic anemia was associated with a high mor-
`tality:rate. ~Most patients would succumb within the
`first six months of the onset of their disease. Treatment
`with anabolic steroids and corticosteroids had,~in most
`cases, been disappointing and most patients died of
`hemorrhagic or infectious complications. The presenting
`symptoms of marrow aplasia are a result of.the, marrow
`failure and are usually easy bruising, spontaneous
`bleeding, and/or infection.
`Severe aplastic anemia is a ~elatively uncommon
`disorder with an inddence of about 65 per million in
`adults over 65 years of age and 4 per:million in children.
`Approximately 25% of cases occur in individuals under
`the age of,20 years and 30% in patieots over the age of
`60 years. Males and females are equally affected. While
`there are well:described congenital forms of marrow
`aplasia including Fanconi~s anemia, the majority of the
`cases are acquired. A variety of etiologic.agents, includ-
`ing chemicals, drugs, ionizing radiation, and infections
`has been implicated. Occasionally pregnancy and thy-
`momas have been. associated with marrow failure, and
`paroxysmal noctttrnal hemoglobinurea may occasionally
`present with pancytopenia. The best known drug as-
`sociation is the rare but often fatal association with
`chloramphenicol. Benzine and insecticides are chemical
`agents that have been implicated in cases of aplastic
`anemia. In spite of the multitude of possible etiologic
`agents, the majority of patients present with no clear-
`cut cause of their marrow failure.
`
`DIAGNOSTIC CRITERIA
`
`The criteria for severe marrow aplasia include a
`markedly hypocellular marrow and peripheral blood
`with any two of the three following findings: ~ neutro-
`phil count of less than 500/dl, a platelet count of less
`than 20,000/dl, and a corrected reticulocyte count of less
`than 1%. Bone marrow aspirations are not adequate to
`make the diagnosis and a bone marrow biopsy is man-
`datory ....
`
`PATHOPHYSIOLOGY
`
`Since aplastic anemia is not a single disease, the
`pathophysiology cannot be explained with a single
`concept. Theoretically, marrow failure could be ex-
`plained as failure of the pleuripotential stem cell, or a
`failure of its microenvironment. Until recently it was
`felt that most cases of marrow failure resulted from an
`isolated failure of the stem cell. HoweVer, clinical ex-
`perience in human marrow transplantation indicates
`that at least in some cases the cause may reside in the
`immune system. There are many-well-documented
`cases of spontaneous autologous’marrow recovery after
`unsuccessful attempts at marrow grafting. In addition,
`at least half of identical twin transplants have failed
`when no immunosuppression was used. Also; a grow-
`ing number of individuals have been successfully
`treated with antithymocyte globulin (ATG). These ob-
`servations implicate an immune mechanism in at least
`some patients.
`
`PHARMACOLOGIC AND IMMUNOLOGIC
`THERAPY
`
`Most of this chapter will deal with bone marrow
`transplantation as the treatment for severe aplastic ane-
`mia; however, a brief description of other forms of
`therapy will also be presented.
`
`STEROIDS
`Treatment with androgens such as testosterone and
`oxymetholone was reported to result in remission of
`severe marrow aplasia in small numbers of patients in
`the early 1960s. These reports were not confirmed in
`lar~ger numbers of patients, and more recent studies in
`the 1970s showed that they provided no advantage over
`modern supportive care. Likewise, corticosteroids in
`291
`
`Sandoz Inc.
`Exhibit 1019-0004
`
`JOINT 1019-0004
`
`

`
`292
`
`V~-BLOOD ANL) NEOPLASTIC DISORDERS
`
`conventional doses (1 to 2 mg/kg) and immunosuppres-
`sive ~trugs such as cydophosphamide have been re-
`ported to be effective in small numbers of patients.
`Confirmation in larger numbers of patients is lacking.
`
`ANTIrHYMOCYTE GLOBULIN
`Since Mathe’s original report on autologous marrow
`recovery in three out of seven patients treated with
`antithymocyte globulin (ATG) in 1970, there have been
`many other studies confirming its efficacy used alone,
`with haplo-identical marrow, or with androgens. The
`doses of ATG and the source of the material have varied
`greatly from study to study. However, the response
`rate in most series is between 40% and 70%. Many
`patients do not respond completely; however, the ma-
`jority of them are transfusion free. The role of haplo-
`identical marrow and androgens remains unclear. The
`use of large doses of methylpredisolone (20 mg/kg)
`along with ATG by the Swiss group has shown re-
`sponses in 15 of 16 patients so treated. In most series
`of patients, the time to response has varied from a few
`weeks to several months and relapses have been seen
`in up to: 10% of ~atients.
`The toxic effect of ATG is considerable and includes
`fever, chills, urticaria, and hypotension. In addition,
`platelet counts usually drop and may be difficult to
`support. Many of these side effects can be modified
`with the use of antihistamines and corticosteroids. Many
`patients will develop serum sickness, which can usually
`be prevented or treated with corticosteroids.
`
`[
`
`Bone Marrow Transplantation
`
`HISTOCOMPA TI BI LITY
`HLA System. Early clinical work, primarily in murine
`systems, ~. showed that marrow could be successfully
`transplanted in lethally irradiated animals only when
`they were histocompatible as determined by the H2
`complex.. The human equivalent is called the human
`leukocyte antigen (HLA) system. There is a series of
`closely related loci on the number six. chromosome
`designated HLA-A, B, C, ahd D, which make up the
`human ’histocompatibility .complex. Because of their
`close proximity on the chromosome, they are inherited
`as a unit in a mendelian codominant manner. Thus
`siblings have a 25% chance of being HLA identical. The
`A, B, C, and DR (D-related) loci are determined,by anti:
`HLA antisera, and the D loci by the mixed lymphocyte
`culture. HLA testing can be done at all transplant
`centers and by most major blood bankS. By and large,
`the majority of successful transplants have been per-
`formed between individuals who are HLA identical. At
`present, a patient must have siblings to have a marrow
`transplant for aplastic anemia. There have been occa-
`sional reports of successful transplants using parental
`or unrelated donors who are phenotypically identical to
`the patient, but these are rare.
`
`PATIENT EVALUATION PRIOR TO
`TRANSPLANTATION
`
`Once the diagnosis has been established, a decision
`needs to be made as to whether the patient is a candi-
`
`date for transplantation. HLA typing of the patient and
`family should be done as soon as possible. The results
`of the HLA typing and mixed lymphocyte culture can
`usually be available within a week. Transplantation in
`patients over the age of 40 years is not usually success-
`ful, and these individuals should be considered for other
`forms of treatment.
`During the evaluation period, blood product trans-
`fusions should be given very judiciously to avoid ex-
`posure of the patient to foreign antigens that might lead
`to graft rejection. Donors who are family members
`should be especially avoided. However, if the patient is
`extremely anemic or bleeding, transfusions should be
`given. The use of washed or frozen red cells and single
`donor, unrelated platelets may help to minimize sensi-
`tization. Infections should be treated vigorously; how-
`ever, it may be necessary to go ahead with the transplant
`even in the face of active infections.
`
`PREPARA TIVE REGIMENS
`Work done~ in animal systems showed ihat some
`form of immunosuppression was necessary in order for
`a marrow transplant to be successful. This has proved
`to be true in the human situation as well. Currently
`most centers, including our own, employ cyclophos-
`phamide as. the backbone of the preparative regimen.
`The dose is 50 :mg/kg of lean body weight given intra-
`venously over one hour daily for four days (total dose
`is 200 mg/kg). Most patients experience intense nausea
`and vomiting with the drug that lasts from several
`hours to several days. Other prominent side effects of
`the drug include an antidiuretic effect, hemorrhagic
`cystitis, and very rarely cardiac toxicity. Because of the
`antidiuretic effect and risk of hemorrhagic cystitis, a
`high urine flow must be maintained. A balanced elec-
`trolyte solution containing sodium bicarbonate, potas-
`sium, and furosemide (10 rag/L) is infused at a rate of
`3000 ml/24 hours/m~. In addition, we use continuous
`bladder irrigation with urologic saline at 1. lite¢ perHour
`beginning at the time of the first cyclophosphamide
`infusion and continuing for 24 to 48 hours after the last
`dose of the drug.
`In patients who have not been Sensitized by prior
`blood product transfusions,i this preparative regimen is
`usually adequate to allow engraftment. However, the
`majority of patients with aplastic anemia have required
`some blood component support either with packed cells
`or with platelets prior to coming ~o transplant. In these
`patients there is a definite risk of graft rejection, which
`may be as high as 30%, and additional immunosuppres-
`sion appears to be necessary. Several methods have
`been used to provide additional immunosuppression.
`Our group currently employs total lymph node irradia-
`tion as suggested by the transplant group at the Uni-
`versity of Minnesota. It involves the delivery of 750 rads
`of irradiation to all the major lymph node-bearing areas
`at a rate of approximately 25 rads per minute. The ~otal
`nodal irradiation is delivered 24 hours after the last dose
`of cyclophosphamide. Other centers have used total
`body irradiation from 300 to 600 rads, and others have
`employed additional drugs such as Myleran and pro-
`carbazine; all of these methods have.been successful in
`reducing the graft rejection rate to less than 10%. The
`Seattle group employs unirradiated donor buffy coat
`transfusions after the marrow transplant for four to five
`days. Recently cyclosporine has been reported by sev-
`
`Sandoz Inc.
`Exhibit 1019-0005
`
`JOINT 1019-0005
`
`

`
`eral European centers to be effective in reducing graft
`rejection,
`
`MARROW PROCUREMENT
`Donors should have a complete physical examina-
`tion, complete blood count, liver and renal function
`tests, electrocardiogram, and chest film prior to being
`accepted. In the event of more than one donor, an
`individua! who is younger, of the same sex, and of the
`same blood group as the patient should be chosen.
`The donor is usually admitted to the hospital a day
`before the procedure. On the rnorning of donation the
`patient is taken to’ the operating room and, under
`general anesthesia, the marrow is aspirated from the
`posterior and, if necessary, anterior iliac crests. During
`the aspirating procedure the marrow is mixed with
`tissue culture media (TC 199, Difco Labs) that contains
`5000 units of preservative-free heparin per 100 ml of
`tissue culture media. The total volume of the marrow
`that is taken will depend on the size and weight of the
`patient and donor, and on the cellularity of the marrow.
`Manual cell counts need to be performed during the
`procedure, and the final cell concentration should be
`greater than 3 x 10-8 nucleated cells per~kg of recipient
`weight. Cel! yields below this are correlated with an
`increased incidence of rejection. Once the marrow has
`been co!lected, it is screened through two sizes of metal
`mesh screens placed on the bottom of cut-off syringes
`and then placed in a standard blood transfer pack. If
`there is no major blood group incompatibility between
`the donor and recipient, the marrow is transfused
`immediately.
`
`POST-TRANSPLANT SUPPORTIVE CARE
`
`ENVIRONMENT
`After the marrow infusion, most patients remain
`pancytopenic for three to four weeks. During this period
`of time, patients should be nursed in single rooms and
`individuals involved with the care of the patient should
`observe careful handwashing techniques. Conventional
`reverse isolation techniques are probably of little value.
`Patients nursed in laminar air flow with gut decontam-
`ination and sterile food may have a decreased incidence
`of graft-Versus-host disease (GVHD) and serious infec-
`tions.
`
`HYPERALIMENTA TION
`Because of infections and oral mucositis, most pa-
`tients are unable to meet their caloric needs orally.
`Hyperalimentation is therefore needed in the majority
`of patients. We use standard hyperalimentation solu-
`tions and attempt to provide 80% to 90% of the patient’s
`caloric needs parenterally. ’ .’
`
`TRANSFUSIONS
`In the majority of patients, adequate blood product
`support can be provided with packed red blood cell
`transfusions and random donor platelet transfusions.
`To avoid the risk of engraftment of lymphocytes in
`these products, all blood products are irradiated with
`2500 rads.
`Occasionally patients will become refractory to ran-
`dora donor platelets and require single donor platelets.
`The marrow donor is usually the best donor in this
`situation, although other falnily members may be used.
`
`It is our practice to a~te:rnpt to keep the hemoglobin
`the range of 10 gm!dl and the platelet count above
`20,000.
`
`TRANSPLANI-RE!_ATED COMPLICATIONS
`
`GRAFT REJECTION
`Failure of engraftment, or graft rejection, is a prob-
`lem that is seen priruarily in marrow transplanfation for
`aplastic anemia. In the early series of patients reported
`by Seattle the rejection rate was approximately 30%,
`and other centers reported rates that were even higher.
`In analyzing their data, taro factors were consistently
`related to graft failure: (1) patients who had been trans-
`fused with blood products prior to undergoing prepa-
`ration for transplantation had a much higher rate of
`rejection than those who were untransfused; and (2)
`those patients who receivedgreater than 3 x 10-~ of
`nucleated cells had a much lower rejection rate. Other
`factors such as patient!donor sex or age have .not proved
`to be of significant prognostic value. Several methods
`have been developed to circumvent this problem and
`they have been alluded to in the section on preparat!ve
`regimens.
`
`INFECTIONS
`Infections are the second major post-transplant
`complication. Many patients with aplastic anemia may
`already be infected at the time they come to transplan-
`tation, and the majority of patients will become clinically
`septic within the first few days after receiving their
`marrow graft. Preventive measures are usually not suc-
`cessful in preventing infections in these profoundly
`immunosuppressed patients. The use of laminar air flow
`in conjunction with gut decontamination with nonab-
`sorbable antibiotics and sterile food and linen has been
`shown to decrease infections but has not improved
`survival.
`
`Management
`The most common organisms that infect patients
`during the first 30 post-t~ansplant days are staphylococ-
`cal species, gram-negative gut flora, and fungi. There-
`fore, when patients become febrile, empiric broad-spec-
`trum antibiotics .should be started. It has been our
`practice to start patients on a semisynthetic penicillin
`such as ticarcillin at 3 gm every four hours, an amino-
`glycoside such as gentamicin or tobramycin at 5
`mg!kg/day in three divided doses, and a cephalosporin
`such as cefazolin at 1 gm every six hours. Peak and
`trougE blood levels of the aminoglycoside should be
`obtained to prevent renal toxic effects and ototoxicity
`and to ensure therapeutic blood levels. Prior to the
`institution of antibiotic therapy, patients should have
`blood, urine and throat cultures obtained. Frequently,
`in spite of the clinical appearance of sepsis, no positive
`cultures will be obtained and the antibiotic choice must
`remain empiric. If the patient continues to be clinically
`septic after the institution of triple antibiotic coverage,
`we add amphotericin B, starting at 0.1 mg/kg/day and
`increasing the dose by 0.1 mg/kg!day up to 0.5 to 0.7
`mg/kg!day; when this dose has been reached, the drug
`is given on an every-other-day schedule: The drug is
`administered over three to four hours. In patients who
`have positive cultures, antibiotic choice can, of course,
`be guided by the sensitivity reports. Patients who have
`
`Sandoz Inc.
`Exhibit 1019-0006
`
`JOINT 1019-0006
`
`

`
`294
`
`persistently positive btood cultures in spite of optimaI~
`antibiotics should be considered for granulocyte trans-
`fusions. To avoid the risk of serious pulmonary reac-
`tions, granulocytes should be separated from ampho-
`tericin by as mucb time as possible. When granulocytes
`are started, they should be continued Until the patient
`is culture negative and the absolute granulocyte count
`is over 500/dl.
`Herpes simplex infections in the ’oral and genital
`regionscan develop during the early post-transplant
`period. The antiviral drug acyclovir given intravenously
`at a dose of 10 mg/kg/dose every eight hours is usually
`effective in suppresging these infections,, Occasionally,
`repeated 10-day courses may be necessary.
`After the granulocyte ~ount has increased to gi~eater
`than 5001dl, the~risk of serious bacterial and fungal
`infections is reduced except in those patients who have
`significant GVHD. These patients often require cortico-
`steroids (2 to 4 mg/kg) and other immun0suppressive
`measures’ to control the. process and, hdnce, ’are at
`continued risk for bacteFial and especially fungal infe&
`lions. In addition, the return of humoral and cellular
`immunity is delayed in these patients, further aggra-
`vating the situation.
`
`INTERSTITIAL PNEUMONIA
`Interstitial pneumonia is a major problem after the
`first 30 post-transplant days. The incidence is lowest in
`those patients who do not receive radiation as part of
`the preparative regimen and in those patients who have
`identical twin (syngeneic) donors. Other risk factors
`include older age and the presence of GVHD. About
`80% of the cases are either idiopathic or related to
`cytomegalovirus (CMV). Rarely other viruses or Pneu.
`mocystis can be the cause of interstitial pneumonia.
`Patients present with steadily increasing hypoxia with
`a paucity of. physical findings. The chest fihn usually
`shows a progressively worsening picture of interstitial
`infiltrates.
`
`Management ~
`Patients with severe GVHD have a very high mor-
`tality rate and to date there has been no effective
`treatment. Antiviral drugs have not.proved effective,
`and the use of high liter immune plasma of globulin
`has not been successful. There has been some early
`evidence that the prophylactic use of high titer CMV
`immune globulin may be effective in preventin, g .the
`development of interstitial pneumonia.
`.... Herpes zoster infections can occur in up to 50% of
`patients following transplantation. The peak incidence
`is at five months. The majority of these patients are still
`very immunocompromised and at risk of cutaneous and
`visceral dissemination.: It is our practice to start ’these
`patients o’n acyclovir in the same doses used for herpes
`simplex infections and to treat for 10 days. ’In most
`patient~ this is adequate treatment. Occasionally, l~a-
`tients will reactivate their disease and require second
`and even third courses of the drug~
`
`G~arr-vr~sus-,osr D,S~ASr (GVHD)
`Acute GVHD. GvHD is the third major complication
`of marrow transplantation. It results from the reaction
`of an immunologically competent donor against an
`
`inm~unologically compromised hos~. The effector cells
`are thought to toe dono>cytotoxic T iymphocytes. Ti~e
`incidence of severe GVHD is between 40% and 70% in
`fully HLA identical allogeneic transplants. The severe
`form is seen in approximately 25% to 30% of patients.
`The target organs are the skin, the gastrointestinal tract,
`and the liver. The clinical manifestations of the disorder
`range from a mild macular skin rash, mild gastrointes-
`tinal discomfort and asymptomatic liver functional
`normalities to a fuhninant and often fatal form with
`~nassive epidermolysis, liver failure, and large quantities
`of diarrhea. Death in these patients is most often from
`intercurrent bacterial, fungal, or viral-infections. A grad-
`ing system for the degree of organ involvement has
`been developed by the Seattle group and is used by
`most transplant centers. Grades 1 and 2 are considered
`the mildest, forms, and grades 3 and 4 are the most
`severe forms, which are associated with the highest
`mortality. Advancing age appears to be the greatest risk
`factor. "
`. Methotrexate given intermittently over the first 100
`post-transplant days has been the most commonly used
`method of attempting to prevent GVHD. It is given at
`15 mg/m~ on the first post-transplant day and at 10
`mg/m~ on day +3, +6, +11, and then weekly there-
`after. Most groups have also added prednisolone at 0.5
`to 2 mg/kg/day to the methotrexate regimen. Antithy-
`mocyte globulin has also been used by some centers
`but the results have been’ inconsistent.
`Cydosporine has been employed by many groups,
`usually starting two to three days prior to marrow
`infusion and continuing for the first three to,six months
`post transplant. However, its place in the prevention
`and treatment of acute GVHD remains to be established.
`Several groups, including our own, have been exploring
`the use of anti-T cell monoclonal antibodies in an
`attempt to remove the T cells from the marrow prior to
`infusion. Preliminary results in fully matched patients
`look promising. Finally, the Seattle group has.reported
`a decreased incidence of grade 2-4 acute GVHD in
`patients cared for in a laminar air flow environment.
`Chronic GVHD. The chronic form of GVHD can
`occur in up to 30% of patients any time after the first
`100 post-transp!an~ g!ays. It may develop as an extension
`of the acute form of the disease, after a quiescent period,
`or de novo. This syndrome may involve multiple organ
`systems including the skin, liver, gastrointestinal tract,
`eyes, lungs, hematopoietic system, and musculoskeletal
`system. The disorder has many of the. characteristics of
`a collagen vascular disease. These patients, as well as
`those with the acute form of the disease have a marked
`delay in the return of their immune function and, hence,
`they are at high risk for developing serious overwhelm-
`ing infections. Most patients suffer from chronic skin
`rashes, hyperpigmentation and hypopigmentation, loss
`of subcutaneous tissues, dry mouth and eyes, chronic
`diarrhea, and liver disease manifested by elevations of
`the transaminases, alkaline phosphatase, and bilirubin.
`Many patients are thrombocytopenic and may have low
`white blood counts, but interestingly, they are not
`usually anemic. Some Suffer from muscle weakness and
`cramps and occasionally joint effusions.
`Treatment has been most successful with the use
`of oral prednisone, usually in doses of I to 2 mg/kg/day.
`Azathioprine in doses of 1.5 mg/kg/day has been used
`
`4
`
`Sandoz Inc.
`Exhibit 1019-0007
`
`JOINT 1019-0007
`
`

`
`by the Seattle group in addition to prednisone and
`initially appeared to improve survival; however, a sub-
`sequent randomized trial has failed to confirm their
`earlier results. The latter patients were treated earlier
`and were usually placed on prophylactic trimethoprim-
`sulfamethoxazole, and received more aggressive sup-
`portive care. In any event, the survival of these patients
`has improved in recent years.
`
`FOLLOW-UP
`
`The length of hospitalization will vary for each
`patient and depend on several factors including the
`rapidity of engraftment, the severity of infections, the
`presence of GVHD and its severity, and the nutritional
`status of the patient. Most patients will spend a total of
`40 to 60 days in the hospital. In younger individuals,
`for those in good general condition prior to undergoing
`transplantation, and in those with few complications,
`the period may be shorter. Once individuals are released
`from the hospital, most require close outpatient follow-
`up for at least two additional months. The patient’s
`graft status and GVHD must be monitored closely. It is
`the practice of most transplant centers to keep patients
`under close observation for the first three months after
`marrow transplantation and then to return them to their
`referring physician.
`
`COST
`
`The cost of the procedure can be quite variable,
`depending primarily on the length of stay in the hospital
`and the number of transplant-related complications that
`the patient experiences. In spite of rising medical costs,
`the cost for most transplants has remained relatively
`constant and is in the range of $75,000 to $!00,000.
`When one compares this with the cost of repeated
`hospitalizations to treat complications of severe aplastic
`anemia, it appears tobe cost-effective.
`Because of its complexity, and the need for highly
`trained medical and paramedical support, this is not a
`procedure that is likely to be easily transported to
`community hospitals. At present it is limited to those
`institutions that have the ne4essary experienced staff
`and facilities to care for these patients.
`
`CLINICAL RESULTS AND CONCLUSIONS
`
`Over the last decade the clinical results for alloge-
`neic marrow transplantation have improved. Long-term
`survival ranges from’40% to around 70%. This can be
`attributed to several factors including earlier transplan-
`tation and better supportive care during the period of
`pancytopenia. The development of methods to prevent
`graft rejection has clearly been the most important factor
`in improving the clinical results in aplastic anemia.
`While a great deal of progress has been made over
`this period of time, GVHD and the lack of a histocom-
`patible donor continue to be major problems in the
`field. If one could obviate GVHD, the limitations of age
`and histocompatibflity could be removed. As discussed
`previously, few patients over the age of 40 are success-
`fully transplanted; this is primarily due to the increased
`
`incidence of GVHD. In fact, the mortality associated
`with the procedure rises rapidly after the age of 20.
`There have been occasional reports of successful
`transplant between less than fully matched patients and
`in a few patients using unrelated fully matched donors.
`However, by and large, these attempts have not been
`successful. Attempts at removing donor T cells from the
`marrow have been successful in fully matched patients,
`but the results in mismatched patients have been dis-
`appointing and clearly much more work needs to be
`done in this area. ’These problems are currently being
`worked on in many marrow transplant centers and it is
`hoped we will see some positive results in the near
`future. Until these problems are worked out, individuals
`who are over the age of 40 and those without histocom-
`patible donors should be considered for treatment with
`alternative forms of therapy such as antithymocyte
`globulin.
`
`REFERENCES
`
`Champlin R, Ho W, Bayever E, ~t al: Treatment of aplastic anemia:
`results with bone marrow transplantation, antithymocyte globulin
`and a monoclonal anti T-cell antibody, h* Young N, Levine A,
`Humphries R: Asplasfic Anemia, Stem Cell Biology and Advances
`in Treatment. Alan R. Liss Inc, New York, 1984, pp 227-238.
`Forman S, Hows J: Bone marrow failure. In Blume K, Petz L (eds):
`Clinical Bone Marrow Transplantation. Churchill-Livingstone, New
`York, 1983, pp 2!5-214.
`Ramsay NKC, Kim T, Nesbit ME, et al: Total lymphoid irradiation and
`cyclophosphamide as preparation for bone marrow transplantation
`in severe aplastic anemia. Blood 55:3442346, 1980.
`Spruce W, McMillan R, Beutler E: Bone marrow transplantation for the
`treatment of severe aplasfic anemia. Clin Hemato112:285-310, 1983.
`Storb R, Thomas ED, Buckner CD, et al: Marrow transplantation for
`aplastic anemia. Semin Hematol 21:27-35, 1984.
`Sullivan K: Graft versus host disease, hi Blume K, Petz L (eds): Clinical
`Bone Marrow Transplantation. Churchill-Livingstone, New York,
`1983, pp 91-120.
`Thomas ED, Storb R: Technique for human marrow grafting. Blood
`36:507-515, 1970.
`Young N, Speck B: Antithymocyte and antilympho~yte globulins: clin-
`ical trials and mechanisms of action. In Young N, Levine A,
`Humphries R: Aplastic Anemia, Stem Cell Biology and Advances
`in Treatment. Alan R. Liss Inc, New York, 1984, pp 221-226.
`Zaia JA: Infections. In Blume K, Petz L (eds): Clinical Bone Marrow
`Transplantation. Churchill-Livingstone, New York, 1983; pp 131-
`!68.
`
`2" IRON DEFICIENCY
`
`Virgil F. Fairbanks
`MAYO CLINIC AND MAYO FOUNDATION
`
`DEFINITIONS AND DIAGNOSTIC CRITERIA
`
`Iron deficiency is the condition in which total body
`iron content is less than normal for a person’s age and
`sex. Three stages of iron deficiency are recognized: (1)
`iron depletion, wherein the normal iron storage pools
`of ferritin and hemosiderin are markedly reduced or
`absent but blood hemoglobin and serum iron concen-
`trations are normal and no physiologic effects of iron
`deficiency are demonstrable; (2) iron deficiency without
`
`Sandoz Inc.
`Exhibit 1019-0008
`
`JOINT 1019-0008
`
`

`
`V 13LOOD AND NEOPLASTIC
`
`~nemia, wherein the iron storage pools are absent,
`serum iron concentration may be reduced, and such
`effects~of iron deficiency as decreased work tolerance or
`epithelial changes may be demonstrated although there
`is no anemia or morphotogic change in erythrocytes;
`and (3) iron deficiency anemia.
`In. general, as iron deficiency progresses, it is re-
`fleeted in the sequence of abnormalities in laboratory
`test results shown in Table 1.
`While Table 1 shows the general pattern in.Which
`changes of iron deficiency evolve, exceptions are com-
`mon. It is not unusual for the serum iron concentration
`(SI), total iron binding capacity (TIBC), and transferrin
`saturation to be normal even when anemia and micro-
`cytosis are evide ~nt..Consequently, these assays are of
`limited diagnosti¢ value, Serum ferritin assay, now
`generally ;avoilable, should supplant assay of SI as a
`diagnostic test. Normal values for serum ferritin vary
`between laboratories, depending on the method used.
`For a commonly used immunoradiometric assay, a value
`of 10 ~g/L or less is usually diagnostic of iron deficiency.
`A major limitation of the ferritin assay is that the serum
`ferritin concentration is usually elevated in the presence
`of chronic "disease, such as rheumatoid arth~iJ~!s, infec-
`tions, and malignancies including lymphoma~ and leu-
`kemias, and this elevation may obscure .concomitant
`iron deficiency.
`Review of a Wright-stain4d blood film, when inter-
`preted in the context of the clinical

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket