throbber
Original article
`
`Annals of Oncology 12: 987-990. 2001.
`© 2001 Kluwer Academic Publishers. Printed in the Netherlands.
`
`Thalidomide in multiple myeloma, myelodysplastic syndromes and
`histiocytosis. Analysis of clinical results and of surrogate angiogenesis
`markers
`
`F. Bertolini,1* W. Mingrone,2* A. Alietti,1 P. F. Ferrucci,1 E. Cocorocchio,1 F. Peccatori,1
`S. Cineri,1 P. Mancuso,1 C. Corsini,1 A. Burlini,1 E. Zucca2 & G. Martinelli'
`1 Division of Hematology-Oncology, European Institute of Oncology, Milan, Italy, 2Oncology Institute of Southern Switzerland. Locarno.
`Switzerland
`
`Summary
`
`Background: Thalidomide, as a single agent, has been recently
`found to induce a clinical response in one third of refractory
`or relapsed myeloma patients. Although it has been reported
`that thalidomide significantly inhibits angiogenesis, it is still
`unclear whether its clinical effect is mediated, at least in part,
`by its anti-angiogenic properties.
`Patients and methods: We evaluated thalidomide as a single
`agent in myeloma, myelodysplastic syndromes (MDS) and
`histiocytosis, i.e. hematological diseases characterized by in-
`creased angiogenesis, and measured prospectively a number of
`surrogate angiogenesis markers.
`Results: Clinical responses were observed in 7 of 17 mye-
`
`loma and 2 of 5 MDS patients. The histiocytosis patient had a
`partial response. At the time of the best clinical response,
`plasma levels of angiogenic growth factors, vascular endothe-
`lial growth factor (VEGF) and basic-fibroblast growth factor
`(b-FGF), were significantly decreased, and flow cytometry
`indicated a decrease of activated endothelial cells in the bone
`marrow of responding MDS patients.
`Conclusions: These observations confirm thalidomide effi-
`cacy in myeloma, suggest a possible use in MDS and histio-
`cytosis and may contribute to the prediction of clinical response
`and to understanding the mechanism of thalidomide's action.
`
`Key words: angiogenesis, histiocytosis. myelodysplastic syn-
`dromes, myeloma, thalidomide
`
`Introduction
`
`Thalidomide, developed in the 1950s as a sedative-
`hypnotic, was withdrawn in the 1960s after reports of
`teratogenicity associated with its use. Thereafter, thali-
`domide has been used for leprosy, discoid lupus ery-
`thematosis, aphthous ulcers in HIV syndromes and
`Behcet's disease [1]. D'Amato et al. [2] have reported
`that, in addition to already known actions (including
`effects on the expression of adhesion molecules, of
`cytokines such as TNF-oc and IL-10, and modulation
`of cell-mediated immunity), thalidomide significantly
`inhibits angiogenesis. Vacca et al. [3] described relevant
`neovascularization in the bone marrow (BM) of mye-
`loma patients, and Singhal et al. [4] reported that thali-
`domide, as a single agent, was able to induce a marked
`and durable response in approximately one third of 84
`chemotherapy-refractory myeloma patients. Although
`two more reports have confirmed the clinical efficacy of
`thalidomide in small series of myeloma patients [5-6],
`it is still unclear whether this effect is mediated, at least
`in part, by its anti-angiogenic properties.
`We have recently reported that angiogenesis is in-
`
`* These authors contributed equally to this work.
`
`creased in myelodysplastic syndromes (MDS), and that
`in this disease BM neovascularization is intermediate
`between healthy controls and acute myeloid leukemia
`[7]. Thus, we decided to evaluate thalidomide as a single
`agent in relapsed myeloma and MDS patients, and to
`measure a number of surrogate markers of angiogenesis
`to gain insight into the biological activity of this drug.
`Here we report an intention-to-treat analysis of 17 mye-
`loma and five MDS patients enrolled in this study and of
`a patient suffering from histiocytosis, another disease
`characterized by increased angiogenesis.
`
`Study design
`
`The median age of the patients was 66 years (range 55-
`80). Of the myeloma patients enrolled, 3 of 17 relapsed
`after tandem high-dose chemotherapy, and 14 of 17
`relapsed after (or were refractory to) at least two lines
`of conventional chemotherapy. MDS patients (two RA,
`three RAEB) failed to respond to cytokine-based thera-
`pies and were treated with supportive care, including red
`cell and platelet transfusions. The histiocytosis patient
`
`DR. REDDY’S LABS., INC. EX. 1046 PAGE 1
`
`

`

`988
`
`was in relapse after a previous velbe-steroid treatment.
`Thalidomide was administered orally at a starting dose
`of 100 mg/day. In the absence of side effects, the dose
`was increased every 2-4 weeks up to 3-400 mg/day.
`Singhal et al. [4] administered thalidomide in a single
`dose in the evening. Similarly to Juliusson et al. [6], and
`taking into account the 6 hours half-life of the drug [8],
`our patients were requested to fractionate the dose in at
`least two daily administrations. Patients were evaluated
`monthly for clinical response and every three months for
`plasma levels of angiogenic growth factors, vascular
`endothelial growth factor (VEGF) and basic-fibroblast
`growth factor (b-FGF), as we previously described [9].
`In MDS patients, the frequency of resting and activated
`BM endothelial cells was evaluated by flow cytometry
`following a procedure that we previously validated in
`preclinical models of human hematopoietic malignan-
`cies [10], and in a clinical study enrolling lymphoma
`and breast cancer patients [11]. All procedures were in
`accordance with the ethical standards of the responsible
`committees on human experimentation and with the
`Helsinki Declaration of the World Medical Association
`
`Results
`
`Side effects were observed in 10 of 23 patients. Three
`patients had constipation, two had Gl-2 neuropathy,
`two had dizziness, one had hypotension. In two patients
`the drug was discontinued for general or gastric intoler-
`ance. Patients without side effects were able to tolerate a
`higher dose of thalidomide. In fact, 7 of 23 patients
`failed to escalate the dose up to 3-400 mg/day because
`of the side effects. On the other hand, split daytime
`dosage did not increase somnolence, fatigue or other
`side effects. One patient reduced the dose to 50 mg/day
`after achieving a clinical response. Overall, the median
`thalidomide dose per day was 220 mg/day (range 50-
`400).
`Of the 17 myeloma patients enrolled, five (29%) had a
`> 50% decrease of the monoclonal Ig, two had a 26%-
`50% reduction, five had a l%-25% reduction, three had
`progressive disease (PD), two discontinued the drug
`within the first 30 days. In responding patients, median
`time to response was 36 days. In this limited group of
`patients, a clear evaluation of dose-response relation-
`ship was not possible. We observed durable clinical
`responses in some patients who achieved a response at
`3-400 mg/day and reduced the drug dosage to 1-200
`mg/day to avoid side effects. Apart from two patients
`who discontinued the drug for side effects, other patients
`discontinued because of progressive disease. Interest-
`ingly, the patient who achieved the best clinical response
`(normalization of Ig levels with 50 mg/day thalidomide
`for more than one year) was the one with highest base-
`line VEGF and b-FGF levels. Of the seven patients with
`a > 25% decrease in monoclonal Ig, three had PD
`(median time to progression 14 months) after a median
`follow-up of 16 months.
`
`(a)
`
`700 -i
`
`(b)
`
`160 -i
`
`Before
`
`After
`
`Before
`
`After
`
`Figure 1. Plasma levels of (a) VEGF and (b) b-FGF before thalido-
`mide administration and at the time of best clinical response in
`myeloma (solid lines), MDS (small dot lines) and histiocytosis (large
`dot line) patients. P = 0.020 and 0 027 by Wilcoxon-matched pair test
`for VEGF and b-FGF. respectively.
`
`Of the five MDS patients, two had a clinical response
`as defined by a recent consensus report [12]. A 64 year-
`old male RAEB patient became transfusion-independent,
`and his BM blasts were reduced from 20% to 5%. Time
`to progression was four months. A 70 year-old female
`RA patient had increased red cell, white cell and platelet
`count, and transfusion interval increased from 15 to 60
`days. Time to progression was seven months. The 63
`year-old female histiocytosis patient had a significant
`decrease of cutaneous lesions and pain, and time to
`progression was six months.
`As shown in Figure 1, at the time of the best response
`both VEGF and b-FGF were significantly decreased
`when compared to pre-treatment values (P = 0.020 and
`0.027 by Wilcoxon matched pair test for VEGF and
`b-FGF, respectively). As reported in Figure 2, flow
`cytometry indicated a striking decrease of activated
`endothelial cells (CD45-, CD14-, CD31+, CD105+) in
`the BM of responding MDS patients.
`
`Discussion
`
`Our data confirm that thalidomide, as a single agent, is
`active in a significant proportion of refractory myeloma
`
`DR. REDDY’S LABS., INC. EX. 1046 PAGE 2
`
`

`

`989
`
`• •;. '.'- , ~b%-/::
`
`..
`
` -
`
`ooo
`
`o>
`
`FSC-Height
`
`1000
`
`FSC-Height
`
`1000
`
`R1
`
`ooo
`
` -
`
`1 °
`
`CD45 PerCP
`
`10
`
`10
`
`10"
`10
`CD4S PerCP
`
`104
`
`o
`
`CO
`
`10"
`10
`CD105FITC
`
`10
`
`o
`
`0.07%
`
`o .
`10° 101
`1
`10
`CD105FITC
`
`)3 104
`
`<D
`
`X6C
`
`O
`CO
`
`Figure 2. Representative dot plots of the frequency of bone marrow-activated endothelial cells evaluated by flow cytometry in a MDS patient
`before (panels on the left) and after (panels on the right) thalidomide Upper panels show the side and the forward scatter of the bone marrow
`cells. A significant Increase of large blasts is observed in the left panel (before thalidomide). whereas a normal distribution is observed after
`therapy. Middle panels show the gate used to exclude CD45+ hematopoietic cells. Panels on the bottom show the frequency of activated
`endothelial cells (CD45-. CD14-. CD31+, CDI05+). A ten fold reduction of activated endothelial cells and return to normal values was observed
`after thalidomide treatment.
`
`patients, and indicate a promising clinical activity of
`single-agent thalidomide in both MDS and histiocyto-
`sis, two hematological malignancies that (similarly to
`myeloma) are associated with relevant angiogenesis.
`Along this line, we observed that circulating angiogenic
`growth factors VEGF and b-FGF, as well as BM acti-
`vated endothelial cells, are significantly decreased at the
`time of best clinical response. It should be noted that,
`unlike serum VEGF, plasma VEGF is not influenced by
`VEGF released from platelets [9, 13]. Thus, fluctuations
`in the platelet count of myeloma and, particularly, MDS
`patients did not bias our measurements of circulating
`VEGF.
`In myeloma [14] and MDS [7], VEGF and b-FGF are
`involved in paracrine loops generated by stroma, endo-
`thelial and neoplastic cells. Thus, our findings support
`the hypothesis that the action of thalidomide might be
`due (at least in part) to the inhibition of cytokine-signal-
`ling between stroma, endothelial and neoplastic cells
`[15]. Sezer et al. [16] recently reported increased VEGF
`and b-FGF levels in myeloma patients compared to
`
`controls. VEGF and b-FGF were found to decrease in
`patients responding to chemotherapy and not in patients
`who did not achieve a remission. Accordingly, the de-
`crease of VEGF and b-FGF observed in the present
`study in myeloma patients at the time of the best
`response to thalidomide might also reflect a decrease in
`tumor load.
`Another relevant issue is whether high levels of
`VEGF, b-FGF and/or BM-activated endothelial cells
`may predict a clinical response to thalidomide. Although
`our series of patients is too small to draw definitive
`conclusions, it should be noted that myeloma patients
`showing higher VEGF and b-FGF levels had clinical
`responses, and that the two MDS patients with higher
`levels of BM-activated endothelial cells responded to
`thalidomide. Singhal et al. [4] reported a decrease in
`BM microvessel density (MVD) in some myeloma pa-
`tients responding to thalidomide, but differences in MVD
`between responding and non-responding patients were
`not statistically significant. Our flow cytometry assay
`generates quantitative data on the frequency of endo-
`
`DR. REDDY’S LABS., INC. EX. 1046 PAGE 3
`
`

`

`990
`
`thelial cells, while MVD indicates the frequency of
`blood vessels. In animal models of human myeloid and
`lymphoid malignancies, tumor engraftment potential,
`speed of engraftment and the frequency of apoptotic
`tumor cells correlated better with the frequency of endo-
`thelial cells enumerated by flow cytometry than with
`MVD evaluation [10]. We are currently enrolling more
`myeloma and MDS patients and evaluating different
`biological parameters to further elucidate the role of
`surrogate angiogenesis markers as prognostic and/or
`predictive factors.
`
`References
`
`1. Thomas DA. Kantarjian HM. Current role of thalidomide in
`cancer treatment. Curr Opin Oncol 2000. 12: 564-73
`2. D'Amalo RJ. Loughnan MS, Flynn E, Folkman J. Thalidomide is
`an inhibitor of angiogenesis. Proc Natl Acad Sci USA 1994; 91:
`4082-5.
`3. Vacca A. Ribatti D, Roncali L et al. Bone marrow angiogenesis
`and progression in multiple myeloma. Br J Haematol 1994; 87:
`503-8.
`4. Singhal S. Mehta J. Desikan R et al. Antitumor activity of
`thalidomide in refractory multiple myeloma. New Engl J Med
`1999:341: 1565-71
`5. Kneller A. Raanani P. Hardan 1 et al. Therapy with thalidomide
`in refractory multiple myeloma patients-the revival of an old
`drug. Br J Haematol 2000; 108: 391-3.
`Juliusson G. Celsing F. Turesson 1 et al. Frequent good partial
`remissions from
`thalidomide including best response ever in
`patients with advanced refractory and relapsed myeloma. Br J
`Haematol 2000; 109. 89-96.
`7. Pruneri G. Bertolini F. Soligo D et al. Angiogenesis in myelodys-
`plastic syndromes Br J Cancer 1999; 81: 1398-401.
`8. Stirling D. Pharmacology of thalidomide. Sem Hematol 2000. 37
`(Suppl 3): 5-14
`
`6
`
`9. Bertolini F. Paolucci M. Peccatori F et al. Angiogenic growth
`factors and endostatin in non-Hodgkin's lymphoma. Br J Hae-
`matol 1999; 106: 504-9.
`10. Fusetti L, Gobbi A, Rabascio C et al. Human myeloid and
`lymphoid malignancies in the NOD/SCID mouse model: Fre-
`quency of apoptotic cells in solid tumors and efficiency and speed
`of engraftment correlate with VEGF production. Cancer Res
`2000; 60: 2527-34.
`11. Mancuso P, Burlini A, Pruneri G et al. Resting and activated
`endothelial cells are increased in the peripheral blood of cancer
`patients. Blood 2001; 97: 3658-61.
`12. Cheson BD Bennett JM; Kantarjian H et al. Report of an
`international working group to standardize response criteria for
`myelodysplastic syndromes. Blood 2000; 96: 3671-4
`13. Wynendaele W. Derua R. Hoylaerts MF et al. Vascular endo-
`thelial growth factor measured in platelet poor plasma allows
`optimal separation between cancer patients and volunteers: A key
`to study an angiogenic marker in v/vo? Ann Oncol 1999; 10:
`965-71.
`14. Dankbar B, Padro T. Leo R et al. Vascular endothelial growth
`factor and interleukin-6 in paracrine tumor-stromal cell inter-
`actions in multiple myeloma. Blood 2000; 95: 2630-6.
`15 Bertolini F, Mancuso P, Gobbi A. Pruneri G The thin red
`line: Angiogenesis in normal and malignant hematopoiesis. Exp
`Hematol 2000; 28: 993-1000.
`16. Sezer O. Jakob C, Eucker J et al. Serum levels of the angiogenic
`cytokines b-FGF, VEGF and HGF in multiple myeloma. Eur J
`Haematol 2001; 66-83-8.
`
`Received 21 February 2001, accepted 11 April 2001.
`
`Correspondence to:
`F. Bertolini. MD. PhD
`Division of Hematology-Oncology
`European Institute of Oncology
`via Ripamonti 435
`20141 Milan
`Italy
`E-mail: francesco.bertolini@ieo.it
`
`DR. REDDY’S LABS., INC. EX. 1046 PAGE 4
`
`

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