throbber
British Journal of Haematology 2000, 109, 89–96
`
`Frequent good partial remissions from thalidomide including
`best response ever in patients with advanced refractory and
`relapsed myeloma
`
`G U N NA R J U L I U S S O N, 1 F R E D R I K C E L S I N G, 2 I N GE M AR T U R E S S O N, 3 S T I G L E NH O F F, 4 M AG N US A D R I A N S S O N
`A ND
`1 1Department of Haematology, University Hospital, Linko¨ping, 2Department of Haematology,
`C L A E S M A L M
`Karolinska Hospital, Stockholm, 3Department of Haematology, University Hospital MAS, Malmo¨,
`4Department of Haematology, University Hospital Lund, and 5Department of Medicine, County Hospital, Kalmar, Sweden
`
`5
`
`Received 16 November 1999; accepted for publication 30 December 1999
`
`Summary. Twenty-three patients with advanced and heavily
`pretreated myeloma were treated with thalidomide. Starting
`dose was 200 mg/d, and 20 patients had dose escalations up
`to 400 (n (cid:136) 5), 600 (n (cid:136) 12) or 800 mg/d (n (cid:136) 3), usually in
`divided doses. Nineteen patients were refractory to recent
`chemotherapy, and four had untreated relapse after prior
`intensive therapy. Ten out of 23 patients (43%) achieved
`partial response (PR; nine with refractory and one with
`relapsed disease), six patients had minor response or
`stabilization of the disease and four had disease progression.
`Another three patients died early from advanced myeloma at
`less than 3 weeks of thalidomide therapy. Of the 10 patients
`with PR, seven had a better response than after any prior
`therapy, despite vincristine–doxorubicin–dexamethasone
`(VAD)-based treatment
`in all but one and high-dose
`melphalan with autologous stem cell support in four. Time
`to achieve PR was rapid in patients receiving thalidomide in
`divided doses
`(median 31 d). Responses also included
`
`reduced bone marrow plasma cell infiltration and improved
`general status. Normalized polyclonal gammaglobulin levels
`were seen in four cases. Six out of 10 patients with PR
`remained in remission with a median time on treatment of
`23 weeks (range 15–50 weeks). Sedation was common but
`usually tolerable, and some patients continued full- or part-
`time work. Four patients had skin problems, three patients
`had pneumonia, one hypothyrosis, one sinus bradycardia
`and one minor sensory neuropathy. Thalidomide may induce
`good partial remissions in advanced refractory myeloma
`with tolerable toxicity, and should be evaluated in other
`settings for myeloma patients. Divided thalidomide doses
`seem to reduce time to achieve remission and may improve
`response rate.
`
`Keywords: myeloma,
`response.
`
`thalidomide,
`
`therapy,
`
`refractory,
`
`Few drug types with documented effect as treatment for
`myeloma are currently available. Steroids and alkylating
`agents, mainly melphalan and cyclophosphamide, have
`significant and dose-related anti-tumour effects. In contrast,
`clinical benefit of anthracyclins have not been documented,
`but still they are frequently incorporated into combination
`chemotherapies (Barlogie et al, 1984). High-dose dexa-
`methasone pulse-based therapy (Barlogie et al, 1984)
`followed by high-dose melphalan with autologous stem cell
`support (Barlogie et al, 1999) leads to a prolonged disease
`control with significant improvement in survival (Attal et al,
`
`Correspondence: Dr Gunnar Juliusson, Department of Haematology,
`University Hospital, SE-581 85 Linko¨ping, Sweden. E-mail: gun-
`nar.juliusson@lio.se.
`
`q 2000 Blackwell Science Ltd
`
`1996; Barlogie et al, 1997; Lenhoff et al, 2000). Still, the
`complete response rate is well below 50%, cure has not
`been documented, as progression-free survival
`is limited
`to a few years, and there is no plateau in the survival
`curves. However, recent studies suggest that allogeneic stem
`cell transplantation may improve response duration and
`outcome.
`Thalidomide (a-(N-phthalimido)glutarimide; C13H10N2O4)
`is a drug that was used as a sedative in the 1960s, but that
`was withdrawn because of severe birth defects. It has a slow
`absorption after oral administration, a plasma half-life of
`about 6 h, and is degraded by non-enzymatic hydrolysis. It
`has immunomodulatory effects with a down-regulation of
`tumour necrosis factor a (Shannon et al, 1997; Haslett et al,
`1998); up-regulation of adhesion molecules (Geitz et al,
`
`89
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 1
`
`

`

`90
`
`G. Juliusson et al
`
`Fig 1. Serum IgG M protein levels for patients
`1–3 (A–C respectively) before and during
`thalidomide treatment. Time points for
`previous therapies are indicated by arrows.
`MP, melphalan 0·25 mg/kg and prednisone
`2 mg/kg for 4 d every 6-weeks; VAD and
`HyperCVAD, given according to Barlogie et al
`(1984) and Dimopoulos et al (1996)
`respectively; SC, autologous stem cell support;
`Cy, cyclophosphamide 2 g/m2; Z (cid:135) TBI (cid:135) M,
`high-dose idarubicin (42 mg/m2), total body
`irradiation (12 Gy) and melphalan 140 mg/
`m2; M iv, melphalan 30–70 mg intravenously;
`M70, melphalan 70 mg/m2; M200, melphalan
`200 mg/m2.
`
`1996) and also antiangiogenetic properties (Bauer et al,
`1998; Or et al, 1998; Berenson et al, 1999; Gasparini, 1999).
`It has been used as therapy for erythema nodosum in leprosy,
`for Behcet’s disease, for HIV-related cachexia and recurrent
`aphthous stomatitis (Jacobsen et al, 1997) and also for
`
`graft-versus-host disease after allogeneic bone marrow
`transplantation (Vogelsang et al, 1992).
`Myeloma is commonly accompanied by an increased
`microvessel density of the bone marrow, which is considered
`to be an adverse prognostic factor (Vacca et al, 1994;
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 2
`
`

`

`Berenson et al, 1999). Such an increased vascularization
`may persist after high-dose therapy (Rajkumar et al, 1999)
`and contribute to relapse.
`Recently, positive treatment results with thalidomide given
`to 84 patients with advanced myeloma were reported as 21
`patients (25%) achieved at
`least partial response from
`thalidomide in doses ranging from 200 to 800 mg per day,
`always given as a single dose in the evening (Singhal et al,
`1999).
`We report here the Swedish experience from advanced
`myeloma treated with thalidomide mostly given in divided
`daily doses.
`
`MATERIALS AND METHODS
`
`Patients. Twenty-three patients with previously treated
`advanced myeloma from five Swedish centres are reported.
`There were 16 men and seven women, and the median age
`was 61·1 years (range 44–78). Myeloma subtype was IgG in
`14 cases, IgA in five and only Bence-Jones proteinuria in
`four. Light chains were kappa in nine, lambda in two and
`data were unavailable in 12. All but one (patient 2; Fig 1)
`had had multiple prior therapies,
`including vincristine–
`doxorubicin–dexamethasone (VAD)-like therapies (Barlogie
`et al, 1984), and 10 had undergone high-dose melphalan
`with autologous stem cell support. The disease duration from
`start of initial treatment was 44 months (range 7–137). At
`inclusion, 19 patients had disease refractory to ongoing
`treatment, whereas four had untested relapse after prior
`intensive therapy. The median plasma cell infiltration in the
`bone marrow was 38% (n (cid:136) 16, range 6–90%), and serum
`beta-2-microglobulin 4·1 mg/l (n (cid:136) 11, range 1·7–5·8). The
`median M-protein level for IgG patients was 49 g/l (range
`21–72 g/l) and for IgA patients it was 24 g/l (range 13–
`44 g/l).
`Drug therapy. Thalidomide tablets (Gru¨ nenthal, Aachen,
`Germany) were given orally, following individual approval
`from the Swedish Medical Drug Agency. The starting dose
`was initially 200 mg twice daily, but as some patients
`became very sedated from the first dose subsequent patients
`started on 100 mg twice daily. Dose escalation to 200 mg
`twice daily was performed within 1 week, and subsequently,
`according to tolerance, to 300 or 400 mg twice daily. Seven
`patients in one centre had one daily dose of up to 800 mg
`thalidomide given in the evening. The dose escalation
`
`Thalidomide in Refractory or Relapsing Myeloma
`
`91
`
`strategy resulted in a maximum thalidomide dose of
`800 mg/d in three patients, 600 mg/d in 12 patients and
`400 mg/d in five; three patients in poor condition who had
`an early death did not have dose escalation. Therapy was
`continued in individual dosing according to response and
`tolerance until prohibitive toxicity, progression, death or
`allogeneic stem cell transplantation. Response was evaluated
`according to the EBMT/IBMTR/ABMTR guidelines (Blade´
`et al, 1998).
`
`RESULTS
`
`Patients with partial response
`Ten patients (43%) had partial response (PR) (Blade´ et al,
`1998) (Tables I and II), two of them had maximum doses of
`800 mg thalidomide/d, seven had 600 mg/d and two had
`400 mg/d. Seven of these patients had a better response to
`thalidomide than to any previous therapy, including high-
`dose melphalan with stem cell support in four and VAD-like
`treatment in all but one (patient 2). PR was achieved in 8/16
`(50%) patients having thalidomide in divided doses, com-
`pared with 2/7 (29%) of those who had single daily doses
`(including patients with early deaths). The clinical courses
`for three of these patients (1–3) are illustrated in Fig 1. One
`patient, who did not tolerate higher doses of thalidomide
`because of somnolence and dizziness, seemed to have a direct
`correlation between thalidomide dose and M-protein level
`(patient 4; Fig 2). Another patient (9) with side-effects due to
`thalidomide had a rapid disease progression after cessation of
`thalidomide. One patient with PR had progression on
`600 mg/d thalidomide after 31 weeks (patient 8), whereas
`other patients have had continuing paraprotein response to
`date or until allogeneic transplantation or death (Table I).
`Time to achieve partial response was rapid; among eight
`patients who had PR to thalidomide in divided doses, the
`median time to PR was 31 d (range 28–81 d), whereas for
`two patients who had PR to single-dose daily therapy
`(patients 7 and 8) the time to PR was 70 d in one and 116 d
`in the other patient. Responding patients rapidly showed
`evidence of responsiveness through declining paraprotein
`levels after only 1 week of therapy (Fig 1). The serum M-
`protein levels before and during thalidomide therapy for all
`individual patients (except for those with early death) are
`shown in Fig 3. Improvements of anaemia and skeletal pain
`was usually slower, although responding patients had a
`
`Table I. Response to thalidomide according to thalidomide dosing (number of patients).
`
`Refractory
`
`Relapsed
`
`Divided/single dose
`
`Divided/single dose
`
`Total
`
`Partial response
`Stable disease/minor response
`Progression
`Early death
`Total
`
`7/2
`1/2
`3/1
`2/1
`13/6
`
`1/0
`2/1
`0/0
`0/0
`3/1
`
`10
`6
`4
`3
`23
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 3
`
`

`

`92
`
`G. Juliusson et al
`
`Table II. Characteristics of patients with partial response to thalidomide.
`
`Patient no.
`(age/sex)
`
`Ig isotype/
`stage
`
`Disease duration
`(months)
`
`Therapy prior to
`thalidomide
`
`Refractory/ relapse
`to prior therapy
`
`Highest dose of
`thalidomide
`
`Thalidomide response
`duration (weeks)
`
`Outcome
`
`1 (57/M)
`
`IgG-kappa/IIIA
`
`10
`
`2 (72/M)
`
`IgG-lambda/IIIA
`
`137
`
`3 (52/M)
`
`IgG/IIIA
`
`4 (56/M)
`
`IgA-kappa/IIIA
`
`5 (54/M)
`
`IgG-kappa/IIIA
`
`6 (44/F)
`7 (54/F)
`8 (64/F)
`
`BJ-kappa/IIIA
`IgG/IIIB
`IgG/IIIA
`
`9 (68/M)
`
`IgG-kappa/IIIA
`
`10 (68/F)
`
`IgA/IIIA
`
`55
`
`51
`
`70
`
`28
`45
`71
`
`34
`
`17
`
`VAD · 3, HyperCVAD · 2,
`M70 (cid:135) SC · 2, M200 (cid:135) SC
`MP · 18
`
`VAD · 3, M200 (cid:135) SC, IFN
`VAD · 2, HDCy, HyperCVAD
`ZTBIM (cid:135) SC, M iv · 5
`VAD · 3, M200 (cid:135) SC, IFN
`VAD · 5, VBAP, VAD · 4, Dex
`VAD · 3, M200 (cid:135) SC
`IFN, VADx2, RT
`VAD · 3, M200 (cid:135) SC, IFN
`VAD · 3, VACP · 5
`CyD (cid:135) IFN · 3, VACP · 24
`
`MP · 5, Z · 3, VAD · 5
`
`CyD · 3, M200 (cid:135) SC
`IFN, RT, MP · 2
`
`Refractory
`
`Refractory
`
`Refractory
`
`Refractory
`
`Refractory
`
`Relapse
`Refractory
`Refractory
`
`Refractory
`
`Refractory
`
`400 mg · 2
`
`200 mg · 2
`
`300 mg · 2
`
`200 mg · 2
`
`300 mg · 2
`
`200 mg · 3
`600 mg · 1
`600 mg · 1
`
`400 mg · 2
`
`300 mg · 2
`
`48
`
`20
`
`32
`
`35
`
`24
`
`20
`50
`31
`
`15
`
`16
`
`Continuing response
`
`Dead, obstructive
`lung disease
`Continuing response
`
`Dose-related
`response
`Allotransplant
`
`Allotransplant
`Continuing response
`Progression on
`treatment
`Progression off
`treatment
`Dead, bone
`marrow failure
`
`IFN, interferon alpha; RT, radiotherapy, CyD, cyclophosphamide (cid:135) dexamethasone; VBAP, vinblastine, BCNU, doxorubicin, prednisone; VACP, etoposide, doxorubicin, cyclophosphamide,
`prednisone; Z, idarubicin orally. For further abbreviations, see legend to Fig 1.
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 4
`
`

`

`Thalidomide in Refractory or Relapsing Myeloma
`
`93
`
`Fig 2. Serum IgA M-protein level and Bence-
`Jones kappa proteinuria excreted per 24 h in
`relation to thalidomide dose in patient 4, who
`experienced dose-related dizziness and vertigo.
`
`significant subsequent improvement of blood counts, symp-
`toms and performance status. Significant improvement of
`cytopenia was seen in patients with minor response.
`Haemoglobin levels and platelet counts from start of
`thalidomide therapy for selected patients with PR or minor
`response are shown in Fig 4.
`Responding patients had a decrease in bone marrow
`plasma cell infiltration to usually below 10%, and serum
`beta-2-microglobulin levels also improved in all five patients
`with follow-up samples available.
`their
`Four responding patients had improvement of
`polyclonal gammaglobulin levels (patients 2, 5, 6 and 10).
`Polyclonal IgG levels were normalized in one patient with
`IgG myeloma (from less than 1 up to 6 g/l; patient 5), one
`with IgA myeloma (4·9 up to 9·8 g/l; patient 10) and one
`with Bence-Jones myeloma (5·2 up to 9·2 g/l; patient 6), and
`one patient with IgG myeloma improved his IgA level from
`0·13 g/l up to 0·93 g/l (patient 2).
`One patient (10), who had a prompt and early reduction of
`her IgA paraprotein during the first month of thalidomide,
`developed a progressive paraparesis due to spinal plasmacy-
`toma that was diagnosed by magnetic resonance tomogra-
`phy and treated with surgery followed by radiotherapy,
`leading to a partial clinical response. She subsequently
`
`developed bone marrow failure 3 months after the start of
`thalidomide and died.
`There was no clear-cut correlation between myeloma
`isotype and response. Patients achieving PR were younger
`(median age 56·5 years vs. 63 years for others), possibly
`reflecting a better ability to accept thalidomide toxicity.
`Nine of 16 (56%) patients refractory to ongoing treatment
`had partial response compared with one of four patients
`given thalidomide as a result of progression off therapy
`(Table I).
`
`Patients with less than partial response
`Among three patients with relapse, minor responses were
`achieved from thalidomide in two, and a significant
`stabilization of the disease in one case. Two of them, both
`with prior autotransplantation, subsequently received oral
`melphalan and prednisone, and had a minor reduction of
`their M-protein levels at the cost of deteriorating blood
`counts. Three patients refractory to previous therapy had
`minor responses, and another had a short clinical improve-
`ment followed by disease progression and death. Three
`patients had continuing myeloma progression,
`in one
`of them thalidomide was discontinued early because of
`skin toxicity.
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`Fig 3. Serum M-protein before and during
`thalidomide treatment for patients with IgG
`and IgA myelomas.
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 5
`
`

`

`94
`
`G. Juliusson et al
`
`Fig 4. Haemoglobin (g/dl) and platelet counts
`(· 109/l) during the first 3 months after the
`start of thalidomide for seven patients with PR
`(thick lines and patient numbers as in Table II)
`and for three patients with minor myeloma
`response (thin lines). Patient 3 received blood
`transfusions during the first month.
`
`Three patients with very advanced disease, one aged 66,
`one aged 68 and one aged 76 years, died within 3 weeks
`from start of
`thalidomide therapy because of disease
`progression without evidence of thalidomide-related toxicity.
`One of them had amyloidosis of the heart.
`
`Toxicity
`All patients became sedated from thalidomide, most sig-
`nificantly within several hours of drug intake. However,
`three of the younger patients managed to continue full-time
`or part-time work. Four patients had to reduce the dose
`because of sedation, vertigo and mood changes. One patient
`did not tolerate thalidomide because of skin toxicity, which
`was reversible but which reappeared on repeated drug
`challenge. Three other patients reported skin problems.
`Three patients had leg oedemas responding to diuretics.
`Moderate obstipation was also observed. One young patient
`had sinus bradycardia with no significant symptoms. One
`elderly patient (2) developed hypothyrosis, which in combi-
`nation with prior chronic obstructive lung disease con-
`tributed to death in good clinical remission from myeloma.
`Two patients with previous recurrent airway infections had
`pneumonia during the first month of thalidomide, and
`another (patient 1) had hypoxia and lung infiltrates with
`cytomegalovirus and Candida albicans diagnosed by bronch-
`eoalveolar lavage. Both patients recovered with treatment
`and had no sequelae. Another patient had pneumonia
`during end-stage disease. One heavily pretreated patient (3;
`Fig 1) had moderate sensory ulnar peripheral neuropathy,
`
`which did not progress during continuing thalidomide
`therapy.
`
`DISCUSSION
`
`We report here the Swedish data on thalidomide therapy for
`advanced myeloma, indicating a high response rate in highly
`refractory disease that was achieved with a low level of
`severe toxicity, although some patients required withdrawal
`or dose reduction because of dose-related toxicities. More
`than half of the patients who were refractory to ongoing
`therapy had partial remission, excluding patients with early
`death due to advanced disease. Of major interest is that many
`of these patients had a better response from thalidomide than
`from any prior therapy,
`including VAD-containing total
`therapy and including high-dose melphalan with autologous
`stem cell support. It is thus clear that thalidomide is an agent
`with major anti-tumour activity against myeloma, and that
`this activity is not cross-reactive to other currently available
`antimyeloma therapies. In fact, in our study, response to
`thalidomide was greater for patients progressing on recent
`chemotherapy than for those with untreated relapse.
`Most of our patients were given a twice daily schedule of
`thalidomide because the plasma half-life of the drug is short
`(< 6 h). This is in contrast to the study by Singhal et al
`(1999),
`in which thalidomide was given once daily. Of
`interest is that in our cohort of patients receiving thalido-
`mide in a single daily dose the time to achieve PR and the
`response rate were similar to that reported by Singhal et al
`(1999), whereas the time to response in those who received
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 6
`
`

`

`thalidomide in divided doses seemed to be improved (Table I).
`The optimal dose and schedule of thalidomide remains to be
`determined.
`Thalidomide has several modes of action, of which the
`antiangiogenetic activity has been suggested to be important
`in myeloma (Bauer et al, 1998; Or et al, 1998; Berenson et al,
`1999; Gasparini, 1999). Increased bone marrow vessel
`density is described and related to response to therapy in
`myeloma (Vacca et al, 1994; Berenson et al, 1999; Rajkumar
`et al, 1999). However, the effect of thalidomide on the
`cytokine network (Jacobsen et al, 1997) might also be
`contributing to the anti-myeloma activity. Both these modes
`of action are less likely for alkylating agents. However, the
`rapid paraprotein response in several of our myeloma
`patients might suggest additional mechanisms, as would
`the rapid progression after withdrawal of thalidomide in two
`others.
`One patient (10) had a rapid major response of her IgA
`paraprotein level, but simultaneously with continuing
`paraprotein improvement she had progressive paraparesis
`due to spinal plasmacytoma. One could speculate whether
`there are differences in the microvessels of the bone marrow
`compartment
`compared with those
`in extraosseous
`tumours, leading to a different response to thalidomide in
`different tissues.
`Although the decline of paraprotein levels was the first
`evidence of response, patients could subsequently benefit
`from improved blood counts and bone marrow findings, but
`most importantly from an improved performance status.
`However, most patients encountered sedation and drowsi-
`ness. In some, these problems enforced withdrawal, but most
`patients tolerated therapy well and some were able to
`continue full- or part-time work. In addition, some patients
`had skin problems that were major in one case and minor in
`others. One patient developed advanced hypothyrosis, which
`might indicate that routine check-ups of thyroid-stimulating
`hormone should be performed. One patient had asympto-
`matic sinus bradycardia, which has been previously reported
`with thalidomide. One patient had moderate peripheral
`neuropathy, which is known to be a significant risk with
`long-term thalidomide therapy. Our case (patient 3) had a
`major general improvement with thalidomide therapy, and
`he had no progression of the neuropathy during continuing
`thalidomide therapy. Of specific interest is that thalidomide
`did not seem to induce bone marrow suppression. Significant
`cytopenia was not seen in this cohort of heavily pretreated
`patients, except for two patients who developed pancytope-
`nia presumed to be myeloma-related during end-stage
`disease. In addition, two younger patients also tolerated
`thalidomide shortly after allogeneic transplantation without
`impairment of their blood counts (patients 5 and 6). On the
`other hand, recovery from cytopenia induced by myeloma or
`by previous treatment was sometimes slow. As regards
`infectious complications, four patients had pneumonia, one
`of which was associated with severe hypoxia and was
`presumably caused by cytomegalovirus and Candida, as
`judged from bronchoalveolar lavage specimens (patient 1).
`All recovered with therapy without sequelae. Thalidomide is
`known to increase HIV virus load in AIDS patients (Jacobsen
`
`Thalidomide in Refractory or Relapsing Myeloma
`
`95
`
`et al, 1997), and it might be considered whether the
`reactivation of cytomegalovirus in our patient was related
`to an immunosuppressive effect of thalidomide. Birth defects
`are rarely if ever a problem in myeloma patients.
`In summary, thalidomide is an old drug with a recently
`identified significant anti-tumour activity in heavily pre-
`treated myeloma patients. Data suggest
`that
`it might
`synergize with conventional chemotherapy, and it does not
`seem to induce bone marrow suppression. Its sedative effects
`are common, but mostly tolerable, and severe side-effects are
`less common and usually reversible. Thus, thalidomide is
`likely to be a major auxiliary drug in myeloma and should be
`evaluated in other settings, such as in combination with
`chemotherapy and in less advanced myeloma.
`
`REFERENCES
`
`Attal, M., Harosseau, J.-L., Stoppa, A.-M., Sotto, J.-J., Fuzibet, J.-G.,
`Rossi, J.-F., Casassus, Ph., Maisonneuve, H., Facon, T., Ifrah, N.,
`Payen, C. & Bataille, R. for the Intergroupe Francaise du Mye´lome.
`(1996) A prospective randomized trial of autologous bone marrow
`transplantation and chemotherapy in multiple myeloma. New
`England Journal of Medicine, 335, 91–97.
`Barlogie, B., Smith, L. & Alexanian, R. (1984) Effective treatment of
`advanced multiple myeloma refractory to alkylating agents. New
`England Journal of Medicine, 310, 1353–1356.
`Barlogie, B., Jagannath, S., Vesole, D.H., Naucke, S., Cheson, B.,
`Mattox, S., Bracy, D., Salmon, S., Jacobson, J., Crowley, J. & Tricot,
`G. (1997) Superiority of tandem transplantation over standard
`therapy for previously untreated multiple myeloma. Blood, 89,
`789–793.
`Barlogie, B., Jagannath, S., Desikan, K.R., Mattox, S., Vesole, D.,
`Siegel, D., Tricot, G., Munshi, N., Fassas, A., Singhal, S., Metha, J.,
`Anaissie, E., Dhodapkar, D., Naucke, S., Cromer, J., Sawyer, J.,
`Epstein, J., Spoon, D., Ayers, D., Cheson, B. & Crowley, J. (1999)
`Total therapy with tandem transplants for newly diagnosed
`multiple myeloma. Blood, 93, 55–65.
`Inhibition of
`(1998)
`Bauer, K.S., Dixon, S.C. & Figg, W.D.
`angiogenesis by thalidomide requires metabolic activation,
`which is
`species-dependent. Biochemical Pharmacology, 55,
`1827–1834.
`Berenson, J.R., Bergsagel, P.L. & Munshi, N. (1999) Initiation and
`maintenance of multiple myeloma. Seminars in Hematology, 36, 9–
`13.
`Blade´, J., Samson, D., Reece, D., Apperley, J., Bjo¨rkstrand, B.,
`Gahrton, G., Gertz, M., Giralt, S., Jagannath, S. & Vesole, D.
`(1998) Criteria for evaluating disease response and progression in
`patients with multiple myeloma treated by high-dose therapy and
`haemopoietic stem cell transplantation. British Journal of Haema-
`tology, 102, 1115–1123.
`Dimopoulos, M.A., Weber, D., Kantarjian, H., Delasalle, K.B. &
`Alexanian, R. (1996) HyperCVAD for VAD-resistant multiple
`myeloma. American Journal of Hematology, 52, 77–81.
`Gasparini, G.
`(1999) The rationale and future potential of
`angiogenesis inhibitors in neoplasia. Drugs, 58, 17–38.
`Geitz, H., Handt, S. & Zwingenberger, K. (1996) Thalidomide
`selectively modulates the density of cell surface molecules involved
`in the adhesion cascade. Immunopharmacology, 31, 213–221.
`Haslett, P.A.J., Corral, L.G., Albert, M. & Kaplan, G.
`(1998)
`Thalidomide costimulates primary human T lymphocytes, pre-
`ferentially inducing proliferation, cytokine production, and
`cytotoxic responses in the CD8(cid:135) subset. Journal of Experimental
`Medicine, 187, 1885–1892.
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 7
`
`

`

`96
`
`G. Juliusson et al
`
`Jacobsen, J.M., Greenspan, J.S., Spritzler, J., Ketter, N., Fahey, J.L.,
`Jackson, J.B., Fox, L., Chernoff, M., Wu, A.W., MacPhail, L.A.,
`Vasquez, G.J. & Wohl, D.A. for the National Institute of Allergy and
`Infectious Diseases AIDS Clinical Trials Group. (1997) Thalido-
`mide for the treatment of oral aphtous ulcers in patients with
`human immunodeficiency virus infection. New England Journal of
`Medicine, 336, 1487–1493.
`Lenhoff, S., Hjorth, M., Holmberg, E., Turesson, I., Westin, J.,
`Nielsen, J.L., Wislo¨ff, F., Brinch, L., Carlsson, K., Carlsson, M.,
`Dahl, I.-M., Gimsing, P., Hippe, E., Johnsen, H., Lamvik, J.,
`Lo¨fvenberg, E., Nesthus, I. & Ro¨djer, S. for the Nordic Myeloma
`Study Group. (2000) Impact on survival of high-dose therapy
`with autologous stem cell support in patients less than 60 years
`with newly diagnosed multiple myeloma: a population-based
`study. Blood, 95, 7–11.
`Or, R., Feferman, R. & Shoshan, S. (1998) Thalidomide reduces
`vascular density in granulation tissue of
`subcutaneously
`implanted polyvinyl alcohol sponges in guinea pigs. Experimental
`Hematology, 26, 217–221.
`
`Rajkumar, S.V., Fonseca, R., Witzig, T.E., Gertz, M.A. & Greipp, P.R.
`(1999) Bone marrow angiogenesis in patients achieving complete
`response after stem cell transplantation for multiple myeloma.
`Leukemia, 13, 469–472.
`Shannon, E.J., Morales, M.J. & Sandoval, F. (1997) Immunomodu-
`latory assays to study structure–activity relationships of thalido-
`mide. Immunopharmacology, 35, 203–212.
`Singhal, S., Mehta, J., Desikan, R., Ayers, D., Roberson, P., Eddlemon,
`P., Munshi, N., Anaissie, E., Wilson, C., Dhodapkar, M., Zeldis, J. &
`Barlogie, B. (1999) Antitumor activity of thalidomide in refractory
`multiple myeloma. New England Journal of Medicine, 341, 1565–
`1571.
`Vacca, A., Ribatti, D., Roncali, L., Ranieri, G., Serio, G., Silvestris, F. &
`Dammacco, F. (1994) Bone marrow angiogenesis and progression
`in multiple myeloma. British Journal of Haematology, 87, 503–508.
`Vogelsang, G.B., Farmer, E.R., Hess, A.D., Altamonte, V., Beschorner,
`W.E., Jabs, D.A., Corio, R.L., Levin, L.S., Colvin, O.M. & Wingard,
`J.R. (1992) Thalidomide for the treatment of chronic graft-versus-
`host disease. New England Journal of Medicine, 326, 1055–1058.
`
`q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 89–96
`
`All in-text references underlined in blue are linked to publications on ResearchGate, letting you access and read them immediately.
`
`DR. REDDY’S LABS., INC. EX. 1060 PAGE 8
`
`

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